EP1575477A2 - LIGAND A FORTE AFFINITE POUR LE RECEPTEUR DE LA NEUROTROPHINE p75 - Google Patents

LIGAND A FORTE AFFINITE POUR LE RECEPTEUR DE LA NEUROTROPHINE p75

Info

Publication number
EP1575477A2
EP1575477A2 EP02729305A EP02729305A EP1575477A2 EP 1575477 A2 EP1575477 A2 EP 1575477A2 EP 02729305 A EP02729305 A EP 02729305A EP 02729305 A EP02729305 A EP 02729305A EP 1575477 A2 EP1575477 A2 EP 1575477A2
Authority
EP
European Patent Office
Prior art keywords
human
pro
amino acid
approximately
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP02729305A
Other languages
German (de)
English (en)
Other versions
EP1575477A4 (fr
EP1575477B1 (fr
Inventor
Barbara L. Hempstead
Ramee Lee
Kenneth K. Teng
Pouneh Kermani
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cornell Research Foundation Inc
Original Assignee
Cornell Research Foundation Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cornell Research Foundation Inc filed Critical Cornell Research Foundation Inc
Publication of EP1575477A2 publication Critical patent/EP1575477A2/fr
Publication of EP1575477A4 publication Critical patent/EP1575477A4/fr
Application granted granted Critical
Publication of EP1575477B1 publication Critical patent/EP1575477B1/fr
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/4045Indole-alkylamines; Amides thereof, e.g. serotonin, melatonin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/185Nerve growth factor [NGF]; Brain derived neurotrophic factor [BDNF]; Ciliary neurotrophic factor [CNTF]; Glial derived neurotrophic factor [GDNF]; Neurotrophins, e.g. NT-3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • A61K38/484Plasmin (3.4.21.7)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/4886Metalloendopeptidases (3.4.24), e.g. collagenase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/55Protease inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the neurotrophins including nerve growth factor (NGF), brain derived neutrophic factor (BDNF), neurotrophin 3 (NT-3), and neurotrophin 4/5 (NT-4/5), are among the molecular determinants that regulate the generation of diverse neuronal populations and the maintenance of their functions within the nervous system (Lewin GR, et al. 1996. Ann. Rev. Neurosci. 19:289; Theonen H. 1995. Science 70:593).
  • trks bind to their cognate receptor tyrosine kinase, trks, (i.e., NGF to trk A, BDNF and NT-4/5 to trk B, and NT-3 to trk C) triggers a cascade of anti-apoptotic events that ensure neuronal survival.
  • trks cognate receptor tyrosine kinase
  • trks i.e., NGF to trk A, BDNF and NT-4/5 to trk B, and NT-3 to trk C
  • the mature form of NGF is more potent than the larger pro-forms of NGF in trk A binding (Chen YE, et al. 1997. Mol. Cell Endocrinol. 127:129) and in promoting cell survival.
  • the p75 receptor belongs to the tumor necrosis factor (TNF) receptor superfamily.
  • TNF tumor necrosis factor
  • the extracellular domain of p75 that is the most critical for neurotrophin binding is reported to be the third and fourth cysteine loops (Choi DW. 1990. J. Neurosci. 10:2493; Yan H, et al. 1991. J. Biol. Chem. 266:12099).
  • the mechanism of signaling elicited by activated trks is unequivocal. However, the molecular and biological consequences of mature neurotrophin-p75 interaction have been confounding.
  • variable loops of mature neurotrophins are most important for interaction with p75 (Ryden M, et al. 1995. Embo J. 14:1979). Despite their importance for binding, these variable loops exhibit the highest degree of variation between the different mature neurotrophins. Nevertheless, all mature neurotrophins bind to p75.
  • apoptosis is believed to be induced in neuronal cells via p75 when mismatching of mature neurotrophin-trk pairing occurs (Majdan M. 1999. IntJ. Dev. Neurosci. 17:153).
  • superior cervical ganglionic neurons express trk A and p75, but not trk B.
  • mature BDNF binds to trk B and to p75, but not to trk A.
  • treatment of superior cervical ganglionic neurons with mature BDNF triggers p75-mediated apoptosis (Bamji SX, et al. 1998. J. Cell Biol. 140:911). Therefore, apoptosis is believed to be induced in neuronal cells when p75 is expressed alone.
  • apoptosis is induced during mismatching of mature neurotrophin-trk pairing or when trk mediated signaling is inhibited.
  • the p75 receptor is up-regulated in neuronal populations after nervous system stress or injury (Roux PP, et al., 1999. J. Neurosci. 19:6887; Widenfalk j, et al. 2001. J Newrosci. 21 :3457; van Eden CG, et al. 1994. Brain Res. Dev. Brain Res. 82:167) and in glia cells of multiple sclerosis patients (Dowling P, et al. 1999. Neurology 53:1676; Chang A, et al. 2000. J. Neurosci. 20:6404). Therefore, under the refined model, activation of p75 by mature neurotrophins in these cells will lead to apoptosis.
  • Recent findings have implicated apoptosis as a common endpoint of various nervous system injuries and environmental insults.
  • Dying neurons in selected instances of, for example, hypoxic ischemia (Choi DW. 1990. J Neurosci. 10:2493), viral infection (Lin KI, et al. 1996. J. Cell Biol. 131 :1149), and neurodegenerative disorders (Roy ⁇ , et al. 1995. Cell 80:167; Liston P, et al. 1996. Nature 379:349) display a number of hallmark characteristics of apoptotic cells.
  • p75 and trks neurotrophins and their receptors
  • p75 expression is distinct from that of the trks (Wheeler EF, et al. 1998. J. Comp Neurol 4:407). The distinct expression supports a trk-independent mechanism for p75 function in non-neuronal cells.
  • NGF and p75 immunoreactivites have been detected in developing incisors (Mitsiadis TA, et al. 1993. Differentiation 54:161).
  • upregulation of p75 in renal biopsies is correlated with various glomerulopathies.
  • the increased expression of p75 detected in renal biopsies apparently recapitulates a normal developing state during kidney development.
  • Both p75 and NGF have been detected in developing muscles.
  • In vitro analysis of the muscle cell line C2C12 suggests that p75-mediated myogenic differentiation is inhibited by NGF.
  • the inhibition of myogenic differention by the NGF-p75 interaction occurs in a trk-independent manner.
  • the expression of p75 is also detected in the atherosclerotic plaques of smooth muscle cells, as well as in many carcinoma cell types.
  • the invention relates to an isolated protein comprising a pro-domain of a proneurotrophin, the pro-domain comprising a pro-domain conserved region, the conserved region being an amino acid sequence having at least approximately 85% identity to amino acid residues approximately at positions 79-98 of human NGF; 91-105 of human BDNF; 101-115 of human NT-3, or 43-57 of human NT-4/5.
  • the invention in another embodiment, relates to a pharmaceutical composition that comprises a protein and a pharmaceutically acceptable additive, the protein comprising: a pro-domain of a proneurotrophin, the pro-domain comprising a pro- domain conserved region, the conserved region being an amino acid sequence having at least approximately 85% identity to amino acid residues approximately at positions 84-98 of human NGF; 91-105 of human BDNF; 101-115 of human NT-3, or 43-57 of human NT-4/5.
  • the invention in another embodiment, relates to an isolated protein comprising: (i) a pro-domain of a proneurotrophin, the pro-domain comprising a pro- domain conserved region; (ii) a mature neurotrophin domain; and (iii) a connector that joins the pro-domain conserved region to the mature neurotrophin domain and that is resistant to protease cleavage.
  • the invention in another embodiment, relates to a pharmaceutical composition that comprises a protein and a pharmaceutically acceptable additive, the protein comprising: (i) a pro-domain conserved region; (ii) a mature neurotrophin domain; and (iii) a connector that joins the pro-domain conserved region to the mature neurotrophin domain and that is resistant to protease cleavage.
  • the invention relates to a nucleic acid molecule that encodes a protein described above.
  • the invention relates to a method for producing a protein described above, the method comprising expressing a nucleic acid molecule that encodes the protein in a host cell.
  • the invention relates to a vector comprising a nucleic acid molecule described above.
  • the invention relates to a host cell comprising a nucleic acid molecule described above.
  • the invention in another embodiment, relates to a method for inducing apoptosis in a cell that comprises p75 receptors on its surface, the method comprising causing the p75 receptor to bind a pharmaceutical composition described above.
  • the invention in another embodiment, relates to a method for cleaving a proneurotrophin protein to a mature neurotrophin, the method comprising contacting the proneurotrophin protein with MMP 3, MMP 7, or plasmin.
  • the invention relates to a method for inhibiting the cleavage of a proneurotrophin to a mature neurotrophin in a mammal in need thereof, the method comprising administering to the mammal an inhibitor of a protease that cleaves the major cleavage site found in native proneurotrophin proteins.
  • the invention relates to a method for preventing or treating atherosclerosis in a mammal, the method comprising administering to the mammal an inhibitor of a protease that cleaves the major cleavage site found in native proneurotrophin proteins.
  • the invention in another embodiment, relates to a method for inducing cell apoptosis in a mammal in need thereof, wherein the cell is susceptible to apoptosis initiated by the binding of a proneurotrophin to a p75 receptor, the method comprising treating the mammal with an inhibitor of a protease that cleaves the major cleavage site found in native proneurotrophin proteins.
  • the invention in another embodiment, relates to a method for inducing apoptosis of cells expressing p75 receptors and trk receptors in a mammal in need thereof, the method comprising administering to the mammal an effective amount of a cleavage-resistant proneurotrophin and an inhibitor of trk activation.
  • the invention in another embodiment, relates to a method for inhibiting apoptosis of a cell in a mammal in need thereof, the method comprising administering to the mammal an effective amount of a molecule that inhibits the binding of a proneurotrophin to a p75 receptor.
  • the invention in another embodiment, relates to a method to screen a human patient for a condition associated with undesired apoptosis, the method comprising determining whether the level of at least one proneurotrophin, or of a conjugate between a p75 receptor and at least one proneurotrophin, in a biological sample of the patient is elevated (i.e. higher than normally occurs in the absence of the condition), wherein elevated levels of proneurotrophins or p75/proneurotrophin conjugates indicate the existence of the condition.
  • the invention in another embodiment, relates to a method for inhibiting apoptosis-mediated hair follicle regression in a mammal in need thereof, the method comprising administering to the mammal an effective amount of a molecule that inhibits the binding of a proneurotrophin to a p75 receptor.
  • Figure 1 A Primary Protein Sequence from Human NGF (GenBank Accession Number: AAA59931);
  • Figure IB Schematic Representation of Monomeric, Unglycosylated Human NGF Isoforms According to Prior Art
  • Figure IC Schematic Representation of Modified Human Pro-NGF According to Invention
  • Figure 2A Primary Protein Sequence from Human BDNF (GenBank Accession Number: AAA69805);
  • Figure 2B Schematic Representation of Monomeric, Unglycosylated Human BDNF Isoforms According to Prior Art
  • FIG. 2C Schematic Representation of Modified Human Pro-BDNF According to Invention
  • Figure 3 A Primary Protein Sequence from Human NT-3 (GenBank Accession Number: AAA59953);
  • Figure 3B Schematic Representation of Monomeric, Unglycosylated Human NT-3 Isoforms According to Prior Art
  • Figure 3C Schematic Representation of Modified Human Pro-NT-3 According to Invention.
  • Figure 4 A Primary Protein Sequence from Human NT-4/5 (GenBank Accession Numbers: AAA60154 & AAA20549)
  • Figure 4B Schematic Representation of Monomeric, Unglycosylated Human NT-4/5 Isoforms According to Prior Art.
  • Figure 4C Schematic Representation of Modified Human Pro-NT-4/5 According to Invention.
  • Figure 4D shows the Human p75 Receptor Sequence (Accession Number: g69056)
  • Figure 5A Alignment results for the human proneurotrophins.
  • the amino acid sequences of human NGF, BDNF, NT-3 and NT-4/5 are aligned for best fit.
  • the signal sequences are boxed (shown as the N-terminal 19-24 amino acids). conserveed positions are shown: * indicates an amino acid conserved in all neutrotrophinis; single and double dots show positions with partial conservation.
  • the mature seqeunces are the longer boxed sequences.
  • Figure 5B Sequence Comparison for NGF across Species.
  • Figure 5C Sequence Comparison for BDNF across Species.
  • Figure 5D Sequence Comparison for NT-3 across Species.
  • Figure 5E Sequence Comparison for NT-4/5 across Species.
  • Figure 6 Purification of His-tagged native (cleaved) NGF, and a His-tagged point mutant (proform) NGF.
  • Media from 293 cells stably expressing constructs of native or mutant NGF inco ⁇ orating a His tag at the carboxy-terminus, or the vector alone were purified using Ni-column chromatography. Proteins eluted with imidazole were subjected to SDS-PAGE and Western blotted with anti-His antisera. Mature (M), and cleavage resistant proform (Pro) are indicated.
  • FIG. 7 Purified pro-NGF (0.8 ⁇ g/ml) was incubated with the indicated proteinases with or without inhibitors and proteolytic products detected by Western blot analysis with anti-NGF antibody.
  • Figure 8 Binding analysis of mature and cleavage-resistant pro-NGF to TrkA or p75 NTR receptors. Cleavage resistant pro-NGF or commercial mature NGF were assayed for their ability to displace 125 I-radiolabeled commercial mature NGF (lnM) from 293 cells expressing TrkA (A) or A875 cells expressing p75 NTR (B). Competition analysis of equilibrium binding was performed using a whole-cell assay. Cells were incubated with 1 nM radioiodinated NGF in the presence or absence of unlabeled mature NGF or unlabeled proNGF from 0.005 to 7 nM concentration for 1 hour at 0.4°C.
  • lnM I-radiolabeled commercial mature NGF
  • Nonspecific binding measured in parallel incubation with 500-fold excess mature NGF was less than 10% of total binding; all results were corrected for this non-specific binding.
  • Each point represents the mean ⁇ standard deviation of quadruplicate determinations. Squares, commercial mature NGF; open circles, cleavage resistant pro-NGF. Competition with purified mature NGF yielded results that were comparable to those obtained with commercial NGF.
  • Figure 9 Distinct biological activities are elicited by cleaved and uncleaved NGF.
  • A Induction of apoptosis by NGF and pro-NGF using p75 expressing smooth muscle cells.
  • Cells expressing p75 were cultured at 39.5°C for 96h to permit differentiation, then incubated with the indicated concentration of commercial NGF (open circles), His-tagged NGF (filled circles), His-tagged pro-NGF (filled squares), or column eluates from vector transfected cells (open squares). After 18h, cells were subjected to TUNEL analysis and were stained with DAPI to visualize nuclei.
  • FIG. 1 Tissue expression of neurotrophins.
  • A NGF
  • B BDNF
  • FIG. 12 Western blot analysis of pro-NGF and pro-BDNF specific antibodies. Recombinant human mature BDNF, or mature mouse NGF, and recombinant cleavage resistant mouse pro-BDNF or recombinant mouse pro-NGF were separated by SDS-polyacrylamide gel electrophoresis. Pro-BDNF was generated by infecting 293 cells with a recombinant adenoviras encoding murine BDNF which had been mutated at positions 434-439. Blots were probed with the indicated antisera to NGF (A) or BDNF (B). Apparent molecular masses are indicated. Figure 13.
  • the present invention is based on the su ⁇ rising discovery by the inventors that uncleaved proneurotrophins bind with higher affinity to p75 than do the cleaved, mature neurotrophins. Moreover, the inventors have discovered that uncleaved proneurotrophins are more effective than mature neurotrophins in inducing apoptosis of cells that express p75, especially cells that express large numbers of p75, or that express p75 exclusively, relative to trk receptors.
  • the p75 receptor is the member of the TNF receptor superfamily that binds all neurotrophins.
  • the p75 receptor has sometimes been called the low affinity neurotrophin receptor.
  • proneurotrophins induce apoptosis of cells via interaction of the proneurotrophins with the p75 receptor. Therefore, it is now, for the first time, apparent that regulated cleavage of proneurotrophins alters their biological activity from promoting death of neuronal cells to promoting survival, and that the proneurotrophins are an integral part of the system. Proteins:
  • one embodiment of the present invention relates to cleavage resistant proteins comprising modifications of native proneurotrophins. Cleavage of such native proneurotrophins generate the family of mature neurotrophins. Proteins comprising modifications of native proneurotrophins in accordance with the invention will be referred to herein as modified or cleavage- resistant proneurotrophins.
  • the native, mature neurotrophin family is well defined, and the members share a number of physical and biological characteristics.
  • biologically active mature neurotrophins are all cleaved from a precursor proneurotrophin at a consensus cleavage site of the furin type (R-X-R/K-R). This cleavage site will be referred to as the major proneurotrophin cleavage site (or, simply, the major cleavage site).
  • the major human proneurotrophin cleavage site includes R-S-K-R in NGF, R-V-R-R in BDNF, R-R-K-R in NT-3, and RSRR in NT-4/5.
  • the mature members of the family interact with the p75 receptor and with the family of trk receptors. All of the mature neurotrophins are involved in the development, survival, and function of neurons in both the peripheral and central nervous systems.
  • the molecular masses of monomeric, unglycosylated mature neurotrophins range from approximately 13.4 to approximately 14 kDa.
  • the isoelectric points of the mature neurotrophins range from approximately 9 to approximately 10.
  • NGF nerve growth factor
  • BDNF brain derived neurotrophic factor
  • NT-3 neurotrophin 3
  • NT-4/5 or NT-4 neurotrophin 4/5 or NT-4.
  • the molecular masses of monomeric, unglycosylated mature NGF, BDNF, NT-3, and NT-4/5 are approximately 13.5, 13.5, 13.6, and 13.9 kDa, respectively.
  • the precursors of the mature neurotrophins i.e. the native proneurotrophins, are also members of a well defined family.
  • the molecular masses of monomeric, unglycosylated proneurotrophins, including the N-terminal signal sequence range from approximately 22 to approximately 30 kDa.
  • the isoelectric points of the proneurotrophins range from approximately 8 to approximately 9.
  • the proneurotrophins are cleaved by proteases at or near the major proneurotrophin cleavage site to produce a mature neurotrophin.
  • pro-NGF pro-BDNF
  • pro-NT-3 pro-NT-4/5.
  • the molecular masses of monomeric, unglycosylated pro-NGF, pro- BDNF, pro-NT-3, and pro-NT-4/5 are approximately 27.0, 27.8, 29.4, and 22.4 kDa, respectively.
  • GenBank accession numbers of human pro-NGF, pro-BDNF, pro- NT-3, and pro-NT-4/5 are AAA59931, AAA69805, AAA59953, and AAA60154/AAA20549, respectively. These sequences are inco ⁇ orated herein by reference, and are displayed in Figures 1A-4A.
  • the present invention is directed to an isolated protein that comprises a pro-domain of a proneurotrophin.
  • the pro-domain optionally further comprises a mature neurotrophin domain and a connector that joins the pro- domain and the mature neurotrophin domain.
  • the definitions of these terms are as described below.
  • pro-domain of a proneurotrophin or, simply “pro-domain” includes all or part of the amino acid sequence at the N-terminus of a native proneurotrophin, as described above, up to, but not including, the major proneurotrophin cleavage site.
  • the pro-domain of a proneurotrophin in accordance with the present invention includes all or part of the following amino acid sequences: amino acid residues 1 to approximately 117 of NGF, 1 to approximately 124 of BDNF, 1 to approximately 134 of NT-3, and 1 to approximately 76 of NT-4/5.
  • the pro-domain comprises at least a pro-domain conserved region, the conserved region being an amino acid sequence having at least approximately 85%, preferably at least approximately 90%, more preferably at least approximately 95% (i.e. one mismatch), and most preferably 100% identity to amino acid residues approximately at positions 79-98 of human NGF; 91-105 of human BDNF; 101-115 of human NT-3, or 43-57 of human NT-4/5.
  • This region will be referred to herein as the pro-domain conserved region, or, simply, the conserved region.
  • the pro-domain of the first embodiment further comprises an amino acid sequence having at least approximately 85%, preferably at least approximately 90%, more preferably at least approximately 95% (i.e. one mismatch), and most preferably 100% identity to amino acid residues approximately at positions 99-117 of human NGF; 106-124 of human BDNF; 116-134 of human NT-3; or 58-76 of human NT-4/5.
  • the pro-domain of the first or second embodiments further comprises an amino acid sequence having at least approximately 85%, preferably at least approximately 90%, more preferably at least approximately 95% (i.e. one mismatch), and most preferably 100%) identity to amino acid residues approximately at positions 66-78 of human NGF; 73-90 of human BDNF; 83-100 of human NT-3; or 25-42 of human NT-4/5.
  • the pro-domain of the third embodiment further comprises an amino acid sequence having at least approximately 85%, preferably at least approximately 90%, more preferably at least approximately 95% (i.e. one mismatch), and most preferably 100%> identity to amino acid residues approximately at positions 46-65 of human NGF; 57-72 of human BDNF; or 61-82 of human NT-3.
  • the pro-domain comprises an amino acid sequence having at least approximately 85%, preferably at least approximately 90%, more preferably at least approximately 95%>, and most preferably 100% identity to amino acid residues approximately at positions 46-117 of human NGF; 57-124 of human BDNF; 61-134 of human NT-3; or 25-76 of human NT-4/5.
  • any of the pro-domains described above optionally comprise a signal sequence, preferably at the N-terminus.
  • the signal sequence characteristically includes a stretch of hydrophobic amino acid residues that facilitates transport into the endoplasmic reticulum, and facilitates the secretion from cells.
  • the pro-domains of the invention may extend to the N-terminus of any of the native proneurotrophins described above.
  • the full length pro-domain may or may not comprise the signal sequence.
  • the invention also includes modified proneurotrophin proteins that are cleavage resistant.
  • the modified proneurotrophins comprise a pro neurotrophin domain, a mature neurotrophin domain, and a connector that joins the pro-domain to the mature domain.
  • the pro-domain may be any of the pro-domains mentioned above.
  • the pro-domain is preferably at the N-terminal end of the sequence, and the mature domain is preferably at the C-terminal end.
  • the mature neurotrophin domain also referred to herein as the mature domain, includes all or part of the amino acid sequence following the major proneurotrophin cleavage site of a native proneurotrophin.
  • the mature domain may comprise all of the amino acid residues of a native, mature neurotrophin protein.
  • the mature neurotrophin domain comprises less than all of the amino acid residues of a native, mature neurotrophin, but at least a sufficient number of amino acid residues to confer on the modified proneurotrophin selective binding to p75, and enhanced apoptosis of cells that express p75, relative to the corresponding mature neurotrophin.
  • the amino acid sequence of a mature neurotrophin domain may comprise at least about 20, 40, 60, 80, or 100 amino acid residues from either the N-terminus of a mature neurotrophin protein, the C-terminus of a mature neurotrophin protein, or anywhere else from within the internal sequence of a mature neurotrophin protein.
  • the mature neurotrophin domain in accordance with the present invention includes all or part of the following amino acid sequences: amino acid residues 122 to 241 of NGF, 129 to 247 of BDNF, 139 to 257 of NT-3, and 81 to 210 of NT-4/5.
  • the modified proneurotrophins of the present invention lack a major proneurotrophin cleavage site.
  • the major cleavage site is replaced by a connector, which is protease cleavage resistant.
  • the connector may be a chemical bond, preferably a covalent or coordinate bond, or any chemical group that is capable of stably joining the pro-domain and the mature domain, and that is protease cleavage resistant.
  • a example of a chemical group useful as a connector is an oligopeptide.
  • the oligopeptide may be any oligopeptide that is protease cleavage resistant, as long as the modified proneurotrophin retains its biological activity, i.e. greater affinity for p75 receptors than mature neurotrophins, and greater capacity to induce apoptosis of cells that express p75.
  • the connector may be a single amino acid, a dipeptide, a tripeptide, a tetrapeptide, a pentapeptide, a hexapeptide or a peptide chain of any length, preferably less than twenty amino acids, more preferably less than ten amino acids.
  • the connector is preferably a tetrapeptide, as is the major proneurotrophin cleavage site. Any amino acid or amino acid sequence may be used in the connector. Some examples of suitable amino acid residues include alanine (A), serine (S), threonine (T), and glycine (G).
  • Any one or more of the amino acid residues in the major proneurotrophin cleavage site may be replaced in order to prevent cleavage.
  • one or all of the dibasic residues, arginine (R) and lysine (K), is replaced.
  • Suitable oligopeptides include A- A, A-S, S-T, P-G, S-S-S, S-G-A, S-A-A, S-A-P, R-S-A-T, G-G-A-P, and S-S-T-P.
  • Particularly favored oligopeptides include the tetrapeptides: R-S-A-A; R-N-A-A; and A-A-A-A.
  • a connector that is "resistant to protease cleavage” means that the connector is substantially uncleavable by a protease that is able to cleave a native proneurotrophin as a result of recognition of the major proneurotrophin cleavage site by the protease.
  • a connector that is resistant to protease cleavage is completely uncleavable by such a protease.
  • proteases that cleave native proneurotrophins include serine proteases.
  • serine proteases are the proprotein convertases, such as furin. Accordingly, the connector is substantially uncleavable, and preferably completely uncleavable, by such serine proteases.
  • MMPs matrix metalloproteinases
  • plasminogen activator proteinases such as tissue plasminogen activator (tPA) and urinary plasminogen activator (uPA).
  • tPA tissue plasminogen activator
  • uPA urinary plasminogen activator
  • the proteins of the invention preferably lack one or more protease cleavage sites in addition to the major cleavage sites, and most preferably lack all protease cleavage sites.
  • sites include, for example, dibasic sites, such as R-R, R-K, K-R, and K-K, as well as other cleavage sites, such as L-L sites.
  • the proteins of the invention comprise all of the isoforms that result from cleavage in any of the pro-domains.
  • the pro-domains comprise isoforms having approximate amino acid residue numbers 1-117, 19-117, 51-117, 81-117, or 82-117 of the human pro-domain of NGF; approximate amino acid residue numbers 1-124, 19-124, 58-124, 110-124, 111-124, or 113-124 of the human pro-domain of BDNF; approximate amino acid residue numbers 1-134, 19-134, or 99-134 of the human pro-domain of NT-3; or approximate amino acid residue numbers 1-76, 25-76, 62-76, or 63-76 of the human pro-domain of NT-4/5.
  • the modified proneurotrophins comprise (i) all or part of a mature domain, (ii) a connector, and (iii) approximate amino acid residue numbers 1-117, 19-117, 51-117, 81-117, or 82-117 of the human pro-domain of NGF; approximate amino acid residue numbers 1-124, 19- 124, 58-124, 110-124, 111-124, or 113-124 of the human pro-domain of BDNF; approximate amino acid residue numbers 1-134, 19-134, or 99-134 of the human pro- domain of NT-3; and approximate amino acid residue numbers 1-76, 25-76, 62-76, or 63-76 of the human pro-domain of NT-4/5.
  • the isoforms described above are derived from cleavage sites present in the pro-domain.
  • the cleavage site may occur approximately four amino acids upstream of the sequence recognition site.
  • proteases that recognize LL sites often cleave within four amino acids upstream of the LL sequence recognition site.
  • the cleavage site may occur approximately four amino acids downstream of the sequence recognition site.
  • the protease furin often cleaves approximately four amino acids downstream of the sequence recognition site. Therefore, the above-mentioned amino acid residue numbers are approximate.
  • the protease cleavage sites described above may be eliminated completely and not replaced, or may be replaced by any of the oligopeptides described above as being useful as connectors.
  • the oligopeptide is preferably a dipeptide, preferably comprising A, S, T, P, or G. Some examples include A- A, T-A, P-T, and G-A.
  • the elimination or replacement of the major proneurotrophin cleavage site, or of one or more additional protease cleavage sites in preparing the proteins of the invention, including the modified proneurotrophins, may be achieved by standard techniques well known in the art. Such techniques include, inter alia, protein synthesis; synthesis and expression of nucleic acid molecules; and site directed mutagenesis and expression of nucleic acid molecules. See, for example, Kolbeck et al., "Characterisation of neurotrophin dimers and monomers," Eur. J. Biochem. 225, 995-1003 (1994). Due to the high conservation of neurotrophin sequences, the pro-domain and the mature domain may be from the same vertebrate or from different vertebrates.
  • the vertebrate may be a non-mammalian vertebrate, such as a frog, fish, bird, or chicken.
  • the vertebrate may be a mammal.
  • the mammal may be a farm animal, such as a goat, horse, pig, or cow; a pet animal, such as a dog or cat; a laboratory animal, such as a mouse, rat, or guinea pig; or a primate, such as a monkey, orangutan, ape, chimpanzee, or human.
  • GenBank accession numbers of sequences of some vertebrate neurotrophins are shown in Table I (A-D) below. The sequences of pro-domains and mature domains are useful in the proteins of the invention, and are inco ⁇ orated by reference herein.
  • sequence of the pro-domain and the sequence of the mature domain may be those of the same proneurotrophin or of different proneurotrophins, independent of whether the sequences are from the same or from a different vertebrate. Accordingly, the pro-domain may have the sequence of any proneurotrophin domain. Similarly, and independently, the mature domain may have the sequence of any mature neurotrophin domain.
  • Some examples of such chimeric modified proneurotrophins include:
  • Pro-NGF-connector-mature BDNF Pro-BDNF-connector-mature BDNF; Pro-BDNF-connector-mature NT-4/5; Pro-NT-3 -connector-mature NGF; Pro-BDNF-connector-mature NT-3; and Pro-BDNF-connector-mature NGF.
  • the proteins of the invention may exist as monomers or multimers.
  • the multimers may comprise any number of proneurotrophin units, such as a dimer, trimer, or tetramer.
  • the proneurotrophin units may be all the same proneurotrophin, in which case the multimer is a homomultimer.
  • the multimer may comprise different proneurotrophin units, in which case the multimer is a heteromultimer.
  • Some examples of heteromultimers include derivatives of NGF - BDGF, NGF-NT-3, BDGF-NT4/5, BDGF-NT-3, and NGF-BDGF-NT4/5.
  • the invention comprises a protein having at least a pro-domain of a proneurotrophin. Any of the pro-domains described above are suitable.
  • the protein may be used as an intermediate to make the modified proneurotrophins described above, or may be used by itself as a substitute for the modified proneurotrophins in the methods of use described below.
  • the protein optionally comprises additional amino acid residues, and may lack a mature domain.
  • the protein further optionally contains a connector, as described above, and/or a major proneurotrophin cleavage site.
  • the pro-domain and the mature domain of the proteins of the invention may further comprise one or more groups commonly associated with amino acid chains.
  • the groups may, for example, result from acetylation, phosphorylation, sulfation, glycosylation or lipidation.
  • the amino acid chain may be chemically bonded to additional peptide and non-peptide moieties.
  • the non-peptide moiety may, for example, be a detectable label, a purification tag, such as a His tag, or a biologically active moiety such as an enzyme or a toxin.
  • the proneurotrophins of the invention include homologs of naturally occuring or native proneurotrophins.
  • a homo log of a native proneurotrophin may be, for example, a substitution mutant, a mutant having an addition or insertion, or a deletion mutant of the protein.
  • Substitutions in a sequence of amino acids are preferably with equivalent amino acids. Groups of amino acids known to be of equivalent character are listed below:
  • Any substitutions, additions, and/or deletions in an amino acid sequence are permitted provided that the protein of the invention continues to satisfy the functional criteria described herein.
  • An amino acid sequence that is substantially identical to another sequence, but that differs from the other sequence by means of one or more substitutions, additions, and/or deletions, is considered to be an equivalent sequence.
  • sequences are aligned so as to maximize the number of identical amino acid residues or nucleotides.
  • sequences of highly homologous proteins and nucleic acid molecules can usually be aligned by visual inspection. If visual inspection is insufficient, the nucleic acid molecules may be aligned in accordance with methods known in the art. Examples of suitable methods include those described by George, D.G. et al., in Macromolecular Sequencing and Synthesis, Selected Methods and Applications, pages 127-149, Alan R. Liss, Inc. (1988), such as formula 4 at page 137 using a match score of 1, a mismatch score of 0, and a gap penalty of -1.
  • less than 15%, more preferably less than 10%, and still more preferably less than 5% of the number of amino acid residues in the sequence of a pro-domain or a mature domain are different (i.e., substituted for, inserted into, or deleted from) from the amino acid residues in the sequence of the corresponding domain of a naturally occuring proneurotrophin to yield the high affinity p75-binding protein of the invention. More preferably still, less than 3%, yet more preferably less than 2% and optimally less than 1% of the number of amino acid residues in a sequence are different from those in a naturally occurring sequence.
  • the proteins of the invention including the modified proneurotrophins of the invention, are isolated.
  • isolated means substantially free from other biological components, as well as from materials that are used in preparation, isolation, characterization or purification of proteins.
  • Some examples of other biological components include cellular components, culture media or components (including conditioned media and components thereof), affinity binding agents, such as immunoconjugates or antibodies and other serum components.
  • materials that are used in preparation, isolation, or purification of proteins include separation media or membranes, such as nitrocellulose, chromatographic matrices, and electrophoretic gel media, including for instance, polyacrylamide and detergents, such as sodium dodecyl sulfate (SDS).
  • the isolated material is at least about 25% to about 90% pure, i.e. free from other proteins and nucleic acid molecules. More preferably, the isolated material is at least about 50% to about 90% pure. Optimally, the isolated material is at least about 75% to about 90% pure.
  • the proteins of the invention are purified.
  • the term "purified” means essentially pure as demonstrated by single band purity on electrophoresis in SDS-polyacrylamide gels (SDS PAGE).
  • the purified material is at least about 90% to about 99.9%) pure. More preferably, the purified material is at least about 95% to about 99.9%) pure. Optimally, the purified material is at least about 99% to about 99.9% pure.
  • Nucleic acids encoding proneurotrophins, vectors, hosts, site directed mutagenesis Nucleic acids encoding proneurotrophins, vectors, hosts, site directed mutagenesis:
  • the invention provides a nucleic acid molecule that encodes any of the proteins mentioned above, such as the pro-domains and the modified proneurotrophin resistant to cleavage by one or more proteases.
  • the sequence of the nucleic acid molecule exists in nature, preferably in a mammalian organism, more preferably, in a human.
  • the encoded protein optionally further comprises a signal sequence and may or may not also include all or part of the mature neurotrophin domain.
  • the nucleic acid may be a DNA molecule, such as for example cDNA or genomic DNA, or alternatively the nucleic acid may be an RNA molecule, such as for example mRNA.
  • the nucleic acid of the invention may be incorporated into a recombinant vector for replication to amplify the nucleic acid, or for expression and isolation/purification of the encoded protein.
  • the recombinant vector may be any recombinant vector, such as a plasmid, a cosmid or a phage.
  • Recombinant vectors have an origin of replication from which copying of the vector and incorporated nucleic acid sequences is initiated.
  • the vector may further include a selectable marker, such as for instance a drug resistance marker, a detectable gene marker or an origin of replication for a second host cell and also a multiple cloning site for ease of manipulation of the inserted nucleic acid.
  • the proteins of the invention are incorporated in pharmaceutical compositions suitable for use as a medicament, for human or animal use.
  • the pharmaceutical compositions may be for instance, in an injectable formulation, a liquid, cream or lotion for topical application, an aerosol, a powder, granules, tablets, suppositories or capsules, such as for instance, enteric coated capsules etc.
  • the pharmaceutical compositions may also be delivered in or on a lipid formulation, such as for instance an emulsion or a liposome preparation.
  • the pharmaceutical compositions are preferably sterile, non-pyrogenic and isotonic preparations, optionally with one or more of the pharmaceutically acceptable additives listed below.
  • compositions of the cleavage resistant proteins of the invention are preferably stable compositions which may comprise one or more of the following: a stabilizer, a surfactant, preferably a nonionic surfactant, and optionally a salt and/or a buffering agent.
  • the pharmaceutical composition may be in the form of an aqueous solution, or in a lyophilized form.
  • the stabilizer may, for example, be an amino acid, such as for instance, glycine; or an oligosaccharide, such as for example, sucrose, tetralose, lactose or a dextram.
  • the stabilizer may be a sugar alcohol, such as for instance, mannitol; or a combination thereof.
  • the stabilizer or combination of stabilizers constitutes from about 0.1 % to about 10% weight for weight of the proneurotrophin.
  • the surfactant is preferably a nonionic surfactant, such as a polysorbate.
  • suitable surfactants include Tween20, Tween ⁇ O; a polyethylene glycol or a polyoxyethylene polyoxypropylene glycol, such as Pluronic F-68 at from about 0.001%) (w/v) to about 10% (w/v).
  • the salt or buffering agent may be any salt or buffering agent, such as for example, sodium chloride, or sodium potassium phosphate, respectively.
  • the buffering agent maintains the pH of the pharmaceutical composition in the range of about 5.5 to about 7.5.
  • the salt and/or buffering agent is also useful to maintain the osmolality at a level suitable for administration to a human or an animal.
  • the salt or buffering agent is present at a roughly isotonic concentration of about 150mM to about 300mM.
  • the pharmaceutical compositions of the present invention may additionally contain one or more conventional additive.
  • additives include a solubilizer such as for example, glycerol; an antioxidant such as for example, benzalkonium chloride (a mixture of quaternary ammonium compounds, known as "quats"), benzyl alcohol, chloretone or chlorobutanol; anaesthetic agent such as for example a morphine derivative; or an isotonic agent etc., such as described above.
  • a solubilizer such as for example, glycerol
  • an antioxidant such as for example, benzalkonium chloride (a mixture of quaternary ammonium compounds, known as "quats"), benzyl alcohol, chloretone or chlorobutanol
  • anaesthetic agent such as for example a morphine derivative
  • an isotonic agent etc. such as described above.
  • the pharmaceutical compositions may be stored under nitrogen gas in vials sealed with impermeable stoppers.
  • the proteins of the invention are useful in methods for the treatment of certain conditions in a mammal.
  • the conditions are mediated by, or involve, cells that express the p75 receptor.
  • the mammal may be a farm animal, such as a goat, horse, pig, or cow; a pet animal, such as a dog or cat; a laboratory animal, such as a mouse, rat, or guinea pig; or a primate, such as a monkey, orangutan, ape, chimpanzee, or human.
  • the invention includes both types of methods.
  • the invention relates to a method for preventing or inhibiting growth of unwanted cells that express p75 receptors.
  • Unwanted cell growth occurs, for example, in malignant cells, such as tumor cells, and in atherosclerotic plaques, especially in the smooth muscle cells of atherosclerotic plaques and vascular ischemias.
  • the malignant cells may, for example, be melanoma cells, lymphoma cells, leukemic cells, prostate cells, pancreatic cells, cells of the testis, lung, brain or heart, or cells of the nervous system, such as cells of the central or peripheral nervous systems.
  • Malignant nervous system cells include malignant neurons and glia cells, such as, for example, medulloblastomas, astrocytomas, and malignant oligodendrocytes.
  • cleavage resistant proteins bind the p75 receptor on the unwanted cells, initiating a commitment to programmed cell death (apoptosis).
  • the invention provides methods of treatment of conditions that are mediated by, or that involve, cells that express the p75 receptor. At least three embodiments of the methods lead to enhanced apoptosis:
  • the first embodiment involves the administration of a protein of the invention in vivo to a human or animal undergoing treatment or otherwise in need thereof.
  • the proteins of the invention either lack a mature domain or are resistant to one or more proteases that would otherwise be effective in the cleavage of the protein to a mature neurotrophin or to a neurotrophin form with residual trk- binding activity.
  • the protein binds the p75 receptor on the surface of cells that are involved in or mediate the condition. The binding of the cleavage resistant proneurotrophin to the p75 receptor induces apoptosis of the cell.
  • the second embodiment involves the administration to a mammal of an inhibitor of a protease that cleaves the major protease cleavage site and/or other cleavage sites of a proneurotrophin.
  • the inhibition reduces or prevents the processing of proneurotrophin to mature neurotrophin.
  • the proneurotrophin binds the p75 receptors on the surface of the cells involved in or mediating the condition, disease or disorder being treated. Binding of the proneurotrophin to the p75 receptors induces apoptosis, as described above.
  • the third embodiment involves a combination of one of the above embodiments with administration of an inhibitor of trk activation.
  • This additional treatment inhibits signaling initiated by any trk receptors that may be present. Activation is initiated by neurotrophin binding and autophosphorylation of the trk receptor.
  • the inhibitor of trk activation blocks trk autophosphorylation, and thereby also blocks subsequent kinase activation in the signal transduction pathways activated by trk.
  • any of the molecules of the invention are capable of inducing p75-mediated apoptosis in vitro and in vivo in both the presence and absence of trk.
  • the methods are especially effective in cells in which trk receptors, if present, are not activated.
  • the level of expression of the p75 receptor may be a normal level of expression, a higher than normal level of expression or even a lower than normal level of expression of p75.
  • a normal level of p75 receptor expression on the surface of a cell is from about 50,000 to about 100,000 such receptors per cell.
  • a higher than normal level of p75 receptor expression includes all cells expressing more than about 100,000 p75 receptors per cell, preferably more than about 150,000 of the p75 receptors per cell; more preferably more than about 200,000 p75 receptors per cell; still more preferably more than about 250,000 p75 receptors per cell and optimally up to about 1,000,000 or more p75 receptors per cell.
  • Cells expressing a lower than normal level of p75 receptors typically have from about 100 to about 50,000 p75 receptors per cell.
  • the invention provides a method for inhibiting activation of the trk receptors of a cell of a mammal in need of such treatment.
  • the method includes the step of administering an effective amount of an inhibitor that inhibits a protease that cleaves the major protease cleavage site of a proneurotrophin.
  • the inhibition of the cleavage of proneurotrophin to mature neurotrophin has the effect of preventing the activation of the cognate trk receptors of the cell by the mature neurotrophin.
  • the protease inhibitor in any of the methods mentioned above may be any inhibitor that inhibits a protease that cleaves the major protease cleavage site of a proneurotrophin.
  • Some classes of proteases include serine proteases, or pro-protein convertases, such as for instance a furin.
  • the protease that cleaves the major protease cleavage site of a proneurotrophin may be plasmin, (activated by a plasminogen activator such as tissue plasminogen activator or urinary plasminogen activator) or a matrix metalloproteinase, such as for instance, MMP-3 or MMP-7.
  • furin inhibitors include, for example, Furin Inhibitor I (Decanoyl-Arg-Nal-Lys-Arg-CMK) and Furin Inhibitor II (hexa-(D) arginine), both of which are available from Calbiochem.
  • plasmin inhibitors include, for example, epsilon amino caproic acid, tramexanic acid, alpha-2 macroglobulin or aprotinin (Se Anes. Anal.2001, 92: 775 and Ann Thor ⁇ c. Surg. 2000, 70: 1300 for details of these inhibitors).
  • inhibitors of matrix metalloproteinases include tetracycline derivatives and other non-peptidic inhibitors such as AG3340 (from Agouron), BAY 12-9566 (from Bayer), BMS-275291 (from Bristol-Myers Squibb) and CGS 27023 A (from Novartis) or the peptidomimetics marimastat and Batimastat (from British Biotech), and the MMP-3 (stromelysin-1) inhibitor, Ac-RCGVPD-NH 2 available from Calbiochem (San Diego, CA). See Hidalgo et al. 2001. J. Natl. Can. Inst. 93: 178-93 for a review of MMP inhibitors in cancer therapy.
  • the invention provides a method for inducing apoptosis of a cell of a mammal in need thereof, the cell being susceptible to initiation of apoptosis by the binding of proneurotrophin to p75 receptors.
  • the method includes the step of administering to the mammal an effective amount of a combination of a protein of the invention along with an inhibitor of trk activation.
  • the inhibitor of trk activation may be any inhibitor of trk activation.
  • Some examples of trk activation include a kinase inhibitor such as for instance, an indolecarbazole derivative.
  • indolecarbazole derivatives include the kinase inhibitor K252a described by Ruggeri et al. 1999. Curr. Med. Chem. 6:845-57.
  • the compound K252a from Cephalon is currently in clinical trials as a candidate for the treatment of pathological conditions of the prostate gland, e.g., benign prostatic hypertrophy or prostate cancer.
  • the invention provides a method for cleaving a proneurotrophin protein to yield a mature neurotrophin.
  • the method comprises contacting the proneurotrophin protein with a particular matrix metalloproteinase. Specifically, the proneurotrophin protein is contacted with MMP-3 or MMP-7. Alternatively, the proneurotrophin protein is contacted with plasmin. Preferably, the proneurotrophin protein is pro-BDNF.
  • the invention provides a pharmaceutical composition comprising a protein of the present invention in combination with an inhibitor that inhibits a protease that cleaves the major protease cleavage site of a proneurotrophin.
  • the combination is preferably in a formulation suitable for use as a medicament for human or animal use as described above.
  • the invention provides a method of treating a condition, disease or disorder mediated by or involving cells with any appreciable level of p75 receptor expression.
  • the method involves administering an effective amount of a pharmaceutical composition comprising the above-recited combination of a protein of the invention in combination with an inhibitor that inhibits a protease that cleaves the major protease cleavage site of a proneurotrophin.
  • the invention provides a method of treating a condition, disease or disorder mediated by or involving cells with any appreciable level of p75 receptor expression, by administering an effective amount of a pharmaceutical composition comprising a protein of the invention in combination with an inhibitor that inhibits trk activation.
  • the invention in another embodiment, relates to a method for inhibiting apoptosis of a cell in a mammal in need thereof.
  • the method comprises inhibiting unwanted binding of a proneurotrophin to a p75 receptor.
  • the proneurotrophin may be any pro-neurotrophin, e.g., pro-NGF, pro-BDNF, pro-NT-3, or pro-NT-4/5.
  • a proneurotrophin refers to an entire proneurotrophin, or to any of the proneurotrophin isoforms that results from cleavage of a pro-domain, such as the isoforms described above.
  • the cell may be any cell that expresses p75, and that undergoes p75-mediated apoptosis.
  • suitable cells include cells of the nervous system, testis, lung, brain, and heart, such as cardiomyocytes.
  • Cells of the nervous system include the various cells of the central nervous system and of the peripheral nervous system.
  • the cells may be neurons or glia cells.
  • Some examples of glia cells include oligodendrocytes, Schwann cells and astrocytes.
  • p75 receptors are present in hair follicles of humans. Typically, the p75 receptors are on keratinocytes and epithelial cells of the outer root sheath.
  • the present invention relates to a method for inhibiting and treating apoptosis-mediated hair follicle regression (e.g. hair loss and baldness) in a human in need thereof.
  • the method comprising administering to the human an effective amount of a molecule that inhibits the binding of a proneurotrophin to a p75 receptor.
  • apoptosis-mediated hair follicle regression e.g. hair loss and baldness
  • the method comprising administering to the human an effective amount of a molecule that inhibits the binding of a proneurotrophin to a p75 receptor.
  • modes of administration may be used in the present embodiment.
  • the preferred mode of administration is topical administration to the skin in the area of the hair follicle regression.
  • the method comprises administering to a mammal a molecule that inhibits the binding of a proneurotrophin to a p75 receptor.
  • a molecule is considered to inhibit the binding of a proneurotrophin to a p75 receptor if the molecule causes a significant reduction in such binding, and the reduction in binding causes a significant reduction in apoptosis.
  • a reduction is considered significant, for example, if the reduction is at least about 10%, preferably at least about 25%, more preferably at least about 75%, and most preferably at least about 90%.
  • the molecule may be any molecule that inhibits the binding of a proneurotrophin to a p75 receptor. In one embodiment, the molecule binds specifically to the p75 receptor, and thereby blocks binding of the proneurotrophin to a p75 receptor, but does not activate the p75 receptor.
  • Some suitable examples of such molecules comprise an antibody hypervariable region that binds specifically to p75 receptors.
  • the hypervariable region may be used alone, or may be part of an entire antibody variable region.
  • the variable region may further comprise an antibody constant region.
  • These molecules may be in the form of antibodies, or any fragment of antibodies, as described above.
  • the molecule that binds specifically to the p75 receptor may also be a small molecule or an oligopeptide.
  • the small molecule or oligopeptide binds specifically to the p75 receptor, blocks binding of the proneurotrophin to a p75 receptor, but does not lead to the biological activity that is caused by binding of the p75 receptor to the proneurotrophin.
  • Such small molecules and oligopeptides can be discovered by methods well known in the art. Typically, discovering such molecules involves providing a cell that expresses p75, providing a small molecule or oligopeptide to be tested, and determining whether the small molecule or oligopeptide to be tested binds to a p75 receptor and, optionally, results in the biological activity caused by binding of a proneurotrophin to a p75 receptor. If the molecule binds with high affinity to the p75 receptor, it is a candidate for use in inhibiting apoptosis. If the molecule binds to the p75 receptor with high affinity and blocks binding of the proneurotrophin to a p75 receptor, it is a stronger candidate. If, in addition to blocking binding, the molecule also fails to cause the biological activity expected from activating a p75 receptor, e.g. apoptosis, the molecule is a candidate for pre-clinical or clinical trials.
  • the oligopeptide has at least approximately four amino acid residues, preferably at least approximately five amino acid residues, and more preferably at least approximately six amino acid residues. The maximum number of amino acid residues is not important, as long as the oligopeptide has the desirable properties mentioned above.
  • the oligopeptide may be linear or cyclic.
  • a cyclic peptide is a linear protein in which a carboxy group, usually the C-terminal carboxy group, forms an amide bond with an amino group, usually the N-terminal amino group.
  • oligopeptides include:
  • Z represents any alpha amino acid and z represents any number from 0 to approximately 20, preferably from 0 to approximately 10, and more preferably from 0 to approximately 5. Any of these oligopeptides may be cyclic.
  • Small molecules include organic compounds, organometallic compounds, salts of organic and organometallic compounds, saccharides, amino acids, and nucleotides. Small molecules typically have molecular weights less than approximately 450 Daltons. Small molecules include compounds that are found in nature as well as synthetic compounds.
  • the present invention includes molecules that bind specifically to the proneurotrophin.
  • the proneurotrophin can be any proneurotrophin, such as the native proneurotrophin molecules identified above.
  • the molecules preferably bind with substantially greater affinity, and preferably bind exclusively, to a proneurotrophin, relative to a mature neurotrophin.
  • molecules that bind specifically to proneurotrophins include molecules that comprise the extracellular domain of a p75 receptor, but that lack the intracellular and trans-membrane domains of a p75 receptor.
  • the extracellular domain may be in a soluble form, or in the form of receptor-Ig chimeras known as receptor bodies or receptobodies.
  • Preferred receptor bodies are divalent homodimers that contain the ligand-binding domain of a receptor followed by the hinge and Fc region of an Ig, such as Igl.
  • the Ig is preferably human Ig.
  • Receptor bodies can be made by methods well known in the art. See, for example, Binder et al. 1999. J. Neurosci. 19:1424-1436 and Marcus et al. 1996. Dev. Biol. 180:786-789.
  • the molecule that binds specifically to the proneurotrophin may comprise an antibody hypervariable region that binds specifically to a proneurotrophin.
  • the hypervariable region may further comprise an entire antibody variable region.
  • the antibody variable region may further comprise an antibody constant region.
  • the molecule that comprises an antibody hypervariable region may be any of the antibodies (including chimerized and humanized antibodies) and antibody fragments (including single chain antibodies) described below.
  • the molecules described above that bind specifically to proneurotrophins may be designed to bind to proneurotrophin monomers or to proneurotrophin multimers.
  • the multimer may be any of the homomultimers and heteromultimers described above.
  • the molecules in the cocktail may comprise receptor bodies, antibodies, or a combination of receptor bodies and antibodies.
  • the cocktail may, for example, comprise one or more molecules that bind specifically to one, two, three, or all of the proneurotrophins.
  • the cocktail may further comprise one or more molecules that binds specifically to the p75 receptor.
  • the molecule that inhibits the binding of a proneurotrophin to a p75 receptor is a protease that cleaves the proneurotrophin to form a mature neurotrophin.
  • the protease may be any of the proteases that cleave proneurotrophins, such as those described above.
  • the mammal in need of inhibiting apoptosis is generally a human who suffers from a condition associated with undesired apoptosis due to binding of a proneurotrophin to a p75 receptor.
  • a condition means any pathological state, such as, for example, a disease, an injury, or any other state that varies from a normal, proper, or healthy state.
  • a condition associated with undesired apoptosis means any condition to which apoptosis contributes either primarily or secondarily, or either directly or indirectly.
  • the condition may involve normal expression in the mammal of the p75 receptor and the proneurotrophin.
  • the condition involves overexpression of the proneurotrophin or of the p75 receptor in the cell undergoing apoptosis, or both.
  • Conditions suitable for treatment in accordance with this embodiment of the invention may, for example, be the result of a nervous system injury or an environmental insult.
  • the condition may, for example, be the result of hypoxic ischemia.
  • Hypoxic ischemia may, for example, be caused by a stroke or by a heart attack.
  • the condition amenable to treatment by the present invention may be caused by a viral infection or a microbial infection.
  • the microbial infection may be a bacterial infection.
  • Some example of conditions caused by a viral infection or a microbial infection include meningitis, encephalitis, or abscesses.
  • neurodegenerative and autoimmune disorders include Alzheimer's Disease, multiple sclerosis, familial dysautonomia, ataxia telangectasia, Charcot-Marie-Tooth disease, Adreno leuko dystrophy, spinal muscular atrophy, Friedriech's ataxia.
  • the condition may also be a condition that causes convulsions.
  • An example of such a condition is epilepsy.
  • an antibody is defined broadly as a protein that binds specifically to an epitope.
  • Antibodies that bind specifically to an epitope may comprise an antibody hypervariable region.
  • the hypervariable region may further comprise an entire antibody variable region.
  • the antibody variable region may further comprise an antibody constant region.
  • the molecule that comprises an antibody hypervariable region may be an antibody including a whole antibody, an antibody fragment, a chimerized antibody or a humanized antibody.
  • the antibody may be polyclonal or monoclonal.
  • Suitable variable and hypervariable regions of non-human antibodies may be derived from antibodies produced by any non-human mammal in which monoclonal antibodies are made. Suitable examples of mammals other than humans include, for example, rabbits, rats, mice, horses, goats, or primates. Preferably, the antibodies are human antibodies. The antibodies may be produced in a transgenic mouse. An example of such a mouse is the so-called XenoMouseTM (Abgenix, Freemont, CA) described by Green, LL., "Antibody Engineering Via Genetic Engineering of the Mouse: XenoMouse Stains are a Vehicle for the Facile Generation of Therapeutic Human Monoclonal Antibodies," J. Immunol Methods," 10;231(1-2):11-23(1999).
  • XenoMouseTM Abgenix, Freemont, CA
  • Antibody fragments have binding characteristics that are the same as, or are comparable to, those of the whole antibody. Suitable fragments of the antibody include any fragment that comprises a sufficient portion of the hypervariable (i.e. complementary determining) region to bind specifically, and with sufficient affinity, to proneutrophins or p75.
  • the preferred fragments are single chain antibodies.
  • Single chain antibodies are polypeptides that comprise at least the variable region of the heavy chain of the antibody and the variable region of the light chain, with or without an interconnecting linker.
  • a chimerized antibody comprises the variable region of a non-human antibody and the constant region of a human antibody.
  • a humanized antibody comprises the hypervariable region (CDRs) of a non-human antibody.
  • the variable region other than the hypervariable region, e.g. the framework variable region, and the constant region of a humanized antibody are those of a human antibody.
  • the antibodies and functional equivalents may be members of any class of immunoglobins, such as: IgG, IgM, IgA, IgD or IgE, and the subclass thereof.
  • the functional equivalents may also be equivalents of combinations of any of the above classes and subclasses.
  • an effective amount of the protein of the invention may be administered to a human or an animal in need thereof by any of a number of well-known methods for use in any of the therapeutic methods described above.
  • the protein may be administered systemically or locally, for example by injection.
  • the systemic administration of a protein of the invention may be by intravenous, subcutaneous, intraperitoneal, intramuscular or intrathecal administration for treatment of a neural cell condition.
  • neural conditions include medulloblastoma, astrocytoma, a malignant dendrocyte or other cancerous condition of a neural cell of either the central nervous system (CNS) or the peripheral nervous system.
  • CNS central nervous system
  • the protein of the invention may be applied topically in appropriate situations.
  • Such situations include, exposed tissue to be treated.
  • exposed tissue include exposed metastatic tissue, as for example in a melanoma.
  • Other exposed cancerous tissue includes cancerous breast tissue.
  • the protein may also be delivered to the scalp of a person to inhibit hair loss due to p75- mediated hair follicle regression.
  • nucleic acid molecules of the invention may be incorporated into recombinant vectors suitable for use in gene therapy.
  • the vector suitable for use in gene therapy may be any vector that comprises a nucleic acid sequence capable of expressing a protein of the invention in a mammal, especially a human, in need of such therapy.
  • the suitable vector may be for example a viral vector, such as an adenovirus vector or an adeno-associated virus (AAV) vector. See for example: Ledley 1996. Pharmaceutical Research 13:1595-1614 and Verma et al. Nature 1997. 387:239-242.
  • nucleic acid molecules of the invention may be administered as naked DNA.
  • the naked DNA may be administered directly to the cells of an organ, tissue or anatomical region most affected by a disorder, condition or disease mediated by or involving cells expressing the p75 receptor.
  • the expressed protein binds to the p75 receptor and induces apoptosis of the cell, thereby preventing any further pathological growth or proliferation of the cell.
  • An effective amount of a pharmaceutical composition of the invention is any amount that is effective to reduce, stabilize or ameliorate the progression of the condition to be treated.
  • the relevant amount usually expressed in mg/kg is determined by routine methods in clinical trials by those of skill in the art.
  • the invention in another embodiment, relates to a method for screening a human patient for a condition associated with undesired apoptosis.
  • the method comprises using a probe to determine the level of at least one proneurotrophin or a proneurotrophin-p75 conjugate in a biological sample.
  • the biological sample can be any sample taken from the human patient.
  • the biological sample may, for example, be blood, urine, hair, cheek scrapings, semen, tissue biopsy, or saliva.
  • the proneurotrophin may be any pro-neurotrophin, e.g., pro-NGF, pro-BDNF, pro-NT-3, or pro-NT-4/5.
  • a proneurotrophin refers to an entire proneurotrophin, or to any of the proneurotrophin isoforms that results from cleavage of a pro-domain. See above.
  • the proneurotrophin-p75 conjugate is any proneurotrophin associated with the p75 receptor.
  • the method comprises determining whether elevated levels of proneurotrophin or proneurotrophin-p75 conjugates are present in the biological sample. The elevated levels indicate the existence of the suspected condition.
  • an elevated level of proneurotrophins means a significantly greater level relative to a human who does not have the condition.
  • a level is significantly greater if the level is at least about 10%, preferably at least about 25%, more preferably at least about 50%, even more preferably at least about 75%o, and most preferably at least about 100% greater than a human who does not have the condition. It is not necessary to determine the level of proneurotrophins in a human who does not have the condition each time.
  • the probe recognizes at least one proneurotrophin or proneurotrophin-p75 conjugate.
  • a probe may, for example, be an antibody, a soluble p75 receptor or a receptor body as described elsewhere in this specification.
  • the probe used to determine the level of at least one proneurotrophin or a proneurotrophin-p75 conjugate in a biological sample may be any molecule that binds specifically to a proneurotrophin or a conjugate.
  • Some examples of probes that recognize proteins include antibodies, soluble receptors or receptor bodies as described elsewhere in this specification.
  • the probe may detect nucleic acid molecules that encode a proneurotrophin.
  • the probe may be an oligonucleotide complementary to the nucleic acid sequence that encodes a proneurotrophin. These oligonucleotide probes are typically designed to detect proneurotrophin mRNA. It should be noted, however, that elevated levels of proneurotrophin mRNA suggest, but are not necessary indicative of, elevated levels of proneurotrophins. The possibility of cleavage of the proneurotrophins into mature neurotrophin must be considered.
  • the probes described above are optionally labelled in accordance with methods known in the art.
  • the label may be a radioactive atom, an enzyme, or a chromophoric moiety.
  • the label may be radioactive.
  • Some examples of useful radioactive labels include 32 P, 125 1, 131 I, 35 S, 14 C, and 3 H. Use of radioactive labels have been described in U.K. 2,034,323, U.S. 4,358,535, and U.S. 4,302,204.
  • non-radioactive labels include enzymes, chromophores, atoms and molecules detectable by electron microscopy, and metal detectable by their magnetic properties.
  • Some useful enzymatic labels include enzymes that cause a detectable change in a substrate.
  • Some useful enzymes and their substrates include, for example, horseradish peroxidase (pyrogallol and o-phenylenediamine), beta-galactosidase (fluorescein beta-D-galactopyranoside), and alkaline phosphatase (5-bromo-4-chloro- 3-indolyl phosphate/nitro blue tetrazolium).
  • horseradish peroxidase pyrogallol and o-phenylenediamine
  • beta-galactosidase fluorescein beta-D-galactopyranoside
  • alkaline phosphatase 5-bromo-4-chloro- 3-indolyl phosphate/nitro blue tetrazolium.
  • Useful chromophores include, for example, fluorescent, chemiluminescent, and bioluminescent molecules, as well as dyes.
  • Some specific chromophores useful in the present invention include, for example, fluorescein, rhodamine, Texas red, phycoerythrin, umbelliferone, luminol.
  • the invention further includes a kit useful in the method for screening a human patient for a condition associated with undesired apoptosis.
  • the kit comprises a probe, and at least one of the following: a label, buffers, standards containing proneurotrophin and its conjugates, materials for developing colorimetric labels, materials for stopping colorimetric reactions, etc.
  • the proteins of the present invention may be prepared by methods that are well known in the art.
  • One such method includes isolating or synthesizing DNA encoding the pro-domains and/or mature domains of the proteins of the invention, and producing the recombinant protein by expressing the DNA, optionally in a recombinant vector, in a suitable host cell.
  • the proteins of the invention may also be made synthetically, i.e. from individual amino acids, or semisynthetically, i.e. from oligopeptide units or a combination of oligopeptide units and individual amino acids. Suitable methods for synthesizing proteins are described by Stuart and Young in “Solid Phase Peptide Synthesis,” Second Edition, Pierce Chemical Company (1984), Solid Phase Peptide Synthesis, Methods Enzymol., 289, Academic Press, Inc, New York (1997) .
  • Nucleic acids encoding the proteins of the invention may also be synthesized in vitro. Suitable methods for synthesizing DNA are described by Caruthers et al. 1985. Science 230:281-285 and DNA Structure, Part A: Synthesis and Physical Analysis of DNA, Lilley, D.M.J. and Dahlberg, J.E. (Eds.), Methods Enzymol., 211. Academic Press, Inc., New York (1992).
  • Nucleic acid molecules encoding the proneurotrophins of the invention may be designed or assembled from known nucleic acid sequences encoding neurotrophins or neurotrophin pro-domains or mature domains.
  • GenBank accession numbers for NGF, BDNF, NT-3, and NT-4/5 are M14805, M61176, M37763, and M86528, respectively.
  • the nucleic acid sequence may be derived from a known proneurotrophin amino acid sequence using the genetic code, as is routine to those of skill in the art.
  • Databases and analytical software for the detection of proteins and nucleic acids having or encoding a particular sequence, pattern or motif are also readily available to the public, and provide useful protein or nucleic acid sequences for adaptation or modification for incorporation into the proneurotrophins of the present invention.
  • NCBI National Center for Biotechnology Information
  • NCBI National Library of Medicine provides databases of protein and nucleic acid sequences and analytical and alignment software at http://www.ncbi.nlm.nih.gov.
  • the proteins of the invention may be isolated from soluble or membrane fractions of preparations, such as for example cell-free lysates, conditioned media or other biological samples containing them by standard methods of protein isolation and purification. Some suitable methods include precipitation and liquid/ chromatographic protocols such as for example, high performance liquid chromatography (HPLC), ion exchange, hydrophobic interaction chromatography, immunoprecipitation, lipid extraction, affinity chromatography and gel filtration to name but a few. See, for example, Guide to Protein Purification, Deutscher, M.P. (Ed.) Methods Enzymol., 182, Academic Press, Inc., New York (1990) and also Scopes, R.K. and Cantor, C.R.
  • HPLC high performance liquid chromatography
  • the nucleic acid encoding the proneurotrophins of the invention may be replicated and expressed in a suitable host cell.
  • suitable host cells include prokaryotic host cells and eukaryotic host cells.
  • Suitable prokaryotic host cells include E. coli cells which are preferred.
  • Suitable eukaryotic host cells include yeast cells, insect cells and mammalian cells, the latter being preferred.
  • Recombinant proneurotrophins are expressed in eukaryotic hosts in preference to prokaryotic hosts in cases where the protein must be post-transcriptionally modified.
  • post- transcriptional modification include glycosylation, phosphorylation, disulfide bond formation, oligomerization and specific cleavage of the transcribed protein product.
  • Prokaryotic hosts do not perform certain post-transcriptional modifications of proneurotrophins of the invention, such as for instance glycosylation. For this reason expression in eukaryotic systems is preferred despite the higher costs associated with production of biologies in eukaryotic systems as compared with the costs of biologies produced in prokaryotic host systems.
  • Prokaryotic host systems are preferred for expression and production of recombinant proneurotrophins of the invention that do not require post-transcriptional modifications that are unique to eukaryotic systems and where the recombinant proneurotrophins are correctly folded or may be refolded in vitro.
  • nucleic acid molecules encoding the proteins of the invention is site directed mutagenesis.
  • Procedures and kits for performing site directed mutagenesis are known in the art. Basically, the nucleic acid sequence of a target sequence within the nucleic acid sequence encoding a native (naturally occurring) proneurotrophin, a chimeric or non-natural proneurotrophin or neurotrophin or fragment of any of the above, may be specifically targeted at will to yield either a random sequence or a predetermined sequence.
  • site directed mutagenesis includes chemical mutagenesis of isolated restriction fragments encoding the target region or sequence, polymerase chain reaction (PCR) mediated mutagenesis of specific sequences and semisynthetic replacement of portions of a coding sequence.
  • PCR polymerase chain reaction
  • Methods for making monoclonal antibodies include, for example, the immunological method described by Kohler and Milstein 1975. Nature 256:495-497 and by Camplbell in "Monoclonal Antibody Technology, The Production and Characterization of Rodent and Human Hybridomas" in Burdon, et al., Eds, Laboratory Techniques in Biochemistry and Molecular Biology, Volume 13, Elsevier Science Publishers, Amsterdam (1985).
  • the recombinant DNA method described by Huse, et al. 1989 Science 246:1275-1281 is also suitable.
  • a host mammal inoculated with an antigen, such as p75 receptor, a proneurotrophin protein, or a proneurotrophin protein-p75 receptor conjugate, as described above, and then, optionally, boosted.
  • an antigen such as p75 receptor, a proneurotrophin protein, or a proneurotrophin protein-p75 receptor conjugate, as described above, and then, optionally, boosted.
  • the receptor fragment must contain sufficient amino acid residues to define the epitope of the molecule being detected. If the fragment is too short to be immunogenic, it may be conjugated to a carrier molecule.
  • Some suitable carrier molecules include keyhole limpet hemocyanin and bovine serum albumin. Conjugation may be carried out by methods known in the art. One such method is to combine a cysteine residue of the fragment with a cysteine residue on the carrier molecule.
  • Spleens are collected from the inoculated mammals a few days after the final boost.
  • Cell suspensions from the spleen are fused with a tumor cell.
  • the resulting hybridoma cells that express the antibodies are isolated, grown and maintained in culture.
  • Suitable monoclonal antibodies as well as growth factor receptor tyrosine kinases for making them are also available from commercial sources, for example, from Upstate Biotechnology, Santa Cruz Biotechnology of Santa Cruz, California, Transduction Laboratories of Lexington, Kentucky, R&D Systems Inc of Minneapolis, Minnesota, and Dako Corporation of Carpinteria, California.
  • Methods for making chimeric and humanized antibodies are also known in the art.
  • methods for making chimeric antibodies include those described in U.S. patents by Boss (Celltech) and by Cabilly (Genentech). See U.S. Patent Nos. 4,816,397 and 4,816,567, respectively.
  • Methods for making humanized antibodies are described, for example, in Winter, U.S. Patent No. 5,225,539.
  • Antibodies or antibody fragments can also be isolated from antibody phage libraries generated using techniques, for example, described in McCafferty et al. 1990. Nature 348: 552-554, using the antigen of interest to select for a suitable antibody or antibody fragment. Clackson et al. 1991. Nature 352: 624-628 and Marks et al. 1991. J. Mol. Biol. 222: 581-597 describe the isolation of murine and human antibodies, respectively, using phage libraries. Subsequent publications describe the production of high affinity (nM range) human antibodies by chain shuffling (Mark et al. 1992. Bio/Technol.
  • CDR-grafting The preferred method for the humanization of antibodies is called CDR- grafting.
  • CDR-grafting the regions of a non-human mammalian antibodies, preferably a mouse antibody, that are directly involved in binding to antigen, the complementarity determining region or CDRs, are grafted into human variable regions to create "reshaped human" variable regions. These fully humanized variable regions are then joined to human constant regions to create complete "fully humanized” antibodies.
  • the human variable regions into which the CDRs will be grafted should be carefully selected, and it is usually necessary to make a few amino acid changes at critical positions within the framework regions (FRs) of the human variable regions.
  • FRs framework regions
  • the reshaped human variable regions may include up to ten amino acid changes in the FRs of the selected human light chain variable region, and as many as twelve amino acid changes in the FRs of the selected human heavy chain variable region.
  • the DNA sequences coding for these reshaped human heavy and light chain variable region genes are joined to DNA sequences coding for the human heavy and light chain constant region genes, preferably 7I and K, respectively.
  • the reshaped humanized antibody is then expressed in mammalian cells and its affinity for its target compared with that of the corresponding murine antibody and chimeric antibody.
  • Methods for making single chain antibodies are also known in the art. Such methods include screening phage libraries transfected with immunoglobulin genes described in U.S. Patent 5,565,332; U.S. Patent 5,5837,242; U.S. Patent 5,855,885; U.S. Patent 5,885,793; and U.S. Patent 5,969,108. Another method includes the use of a computer-based system for designing linker peptides for converting two separate polypeptide chains into a single chain antibody described in U.S. Patent 4,946,778; U.S. Patent 5,260,203; U.S. Patent 5,455,030; and U.S. Patent 5,518,889.
  • the labels may be conjugated to probes by methods that are well known in the art.
  • the labels may be directly attached through a functional group on the probe.
  • the probe either contains or can be caused to contain such a functional group.
  • suitable functional groups include, for example, amino, carboxyl, sulfhydryl, maleimide, isocyanate, isothiocyanate.
  • labels such as enzymes and chromophoric molecules may be conjugated to the antibodies or nucleotides by means of coupling agents, such as dialdehydes, carbodiimides, dimaleimides, and the like.
  • the label may also be conjugated to the probe by means of a ligand attached to the probe by a method described above and a receptor for that ligand attached to the label.
  • a ligand attached to the probe by a method described above and a receptor for that ligand attached to the label.
  • Any of the known ligand-receptor combinations is suitable.
  • Some suitable ligand-receptor pairs include, for example, biotin-avadin or -streptavadin, and antibody-antigen.
  • the biotin-avidin combination is preferred.
  • the probe may be an antibody, preferably a monoclonal antibody.
  • the antibodies may be prepared as described above.
  • the antibodies described above can be used in assays to detect the presence of the proteins, conjugates, and complexes.
  • assays can be performed using known formats such as immunohistochemistry/immunocytochemistry of tissues (U.S. Patent No. 5846749) and ELISA (Current Protocols in Immunology, Wiley Intersciences, New York, 1999).
  • Assays for detecting the presence of proteins with antibodies have been previously described, and follow known formats, such as standard blot and ELISA formats. These formats are normally based on incubating an antibody with a sample suspected of containing the protein and detecting the presence of a complex between the antibody and the protein. The antibody is labelled either before, during, or after the incubation step.
  • the protein is preferably immobilized prior to detection. Immobilization may be accomplished by directly binding the protein to a solid surface, such as a microtiter well, or by binding the protein to immobilized antibodies.
  • a protein is immobilized on a solid support through an immobilized first antibody specific for the protein.
  • the immobilized first antibody is incubated with a sample suspected of containing the protein. If present, the protein binds to the first antibody.
  • a second antibody also specific for the protein, binds to the immobilized protein.
  • the second antibody may be labelled by methods known in the art. Non- immobilized materials are washed away, and the presence of immobilized label indicates the presence of the protein.
  • This and other immunoassays are described by David, et al. in U.S. Patent 4,376,110 assigned to Hybritech, Inc., La Jolla, California; by Coligan, J.E, et al. (Eds.), Current Protocols in Immunology, Wiley Intersciences, New York, 1999); and by Harlow, E. and Lane, D., Using Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1999).
  • Immunoassays may involve one step or two steps.
  • the target molecule if it is present, is immobilized and incubated with a labelled antibody.
  • the labelled antibody binds to the immobilized target molecule.
  • the sample is assayed for the presence of the label.
  • immobilized target molecule is incubated with an unlabelled first antibody.
  • the target molecule-antibody complex if present, is then bound to a second, labelled antibody that is specific for the unlabelled antibody.
  • the sample is washed and assayed for the presence of the label, as described above.
  • immunometric assays described above include simultaneous sandwich, forward sandwich, and reverse sandwich immunoassays. These terms are well known to those skilled in the art.
  • the sample containing antigen, solid phase immunoabsorbent with immobilized antibody and labeled soluble antibody are incubated under conditions and for a period of time sufficient to allow antigen to bind to the immobilized antibodies and to the soluble antibodies.
  • the specific concentrations of labeled and immobilized antibodies, the temperature and time of incubation, as well as other such assay conditions can be varied, depending upon various factors including the concentration of antigen in the sample, the nature of the sample and the like. Those skilled in the art will be able to determine operative and optimal assay conditions for each determination by employing routine experimentation.
  • solid phase immunoabsorbents which have been employed and which can be used in the present invention.
  • Well known immunoabsorbents include beads formed from glass, polystyrene, polypropylene, dextran, nylon, and other material; and tubes formed from or coated with such materials, and the like.
  • the immobilized antibodies may be covalently or physically bound to the solid phase immunoabsorbent, by techniques such as covalent bonding via an amide or ester linkage or by absorption.
  • Example 1 Expression and isolation of His-tagged cleavage resistant NGF:
  • NGF cDNAs were stably expressed in 293 cells, to generate microgram quantitites of His-tagged native NGF, which is completely processed to the mature form of 13.5 kDa in 293 media and a His-tagged point mutant of pro-NGF, in which the K-R cleavage site at 120,121 has been mutated to A-A. This renders the molecule cleavage resistant, and results in a proform of NGF of 30 kDa isolated from 293 cell media (Fig. 6).
  • proNGF secreted proNGF
  • purified proNGF was incubated with the proteinases with or without inhibitors.
  • Pro-NGF was cleaved by plasmin to a 13 kD form.
  • Inhibition of plasmin activity upon addition of aprotinin inhibited cleavage of pro-NGF.
  • incubation of the pro-NGF with MMP-7 but not MMP-2, -3, or -9 resulted in cleavage of the 30 kD proform of NGF to the 17 kD form (Fig. 7).
  • Example 3 Assays of the biological activity of cleavage resistant NGF:
  • binding assays (2) apoptosis assays using cells expressing only p75 and (3) assays of trk activity, using trk-mediated autophosphorylation in dose-response studies and PC 12 cell neuritogenesis.
  • the cleavage resistant proform of NGF binds with higher affinity than the cleaved mature form of NGF to the p75 receptor.
  • Competition binding assays were performed to determine the affinity of interaction of the uncleaved proform to p75 or to trk A.
  • Example 4 The cleavage resistant pro-NGF is more effective than mature NGF in inducing apoptosis of cells expressing only p75:
  • a vascular smooth muscle cell line expressing p75 (but not trk receptors) which exhibits dose dependent apoptosis upon NGF addition was used in the following experiments.
  • Cells were exposed to His tagged mature NGF or His tagged uncleaved NGF purified from 293 cell media (see Fig. 6) or as controls, purified media from cells transfected with vector or commercial (predominantly mature) NGF.
  • Apoptosis was assessed by TUNEL analysis at 18h post-treatment (See Fig. 9 A). Less than 5% of the control cells were TUNEL- positive.
  • Example 5 The cleavage resistant pro-NGF is less effective in activating trk:
  • Example 6 The cleavage resistant pro-NGF is less effective than mature NGF in mediating trk dependent neuritogenesis:
  • PC 12 cells were utilized in neuritogenesis assays (Fig. 9B). Treatment of these cells with His-tagged mature, or commercial (predominantly mature) NGF, induced neurites at 0.1 nM concentrations. However, no neurite outgrowth was observed in cells that were treated with the cleavage resistant pro-NGF even at concentrations up to 2 nM. PC 12 cells treated with purified media from cells transfected with vector displayed no neurite outgrowth. As a control, TUNEL analysis was performed on replicate cultures; less than 2%> cell death were observed under all conditions tested.
  • Example 7 Pro-NGF induces cell death in neurons coexpressing p75 and trk A:
  • Superior cervical ganglia neurons coexpress both p75 and trk A receptors.
  • the neurons were treated with either mature NGF or pro-NGF.
  • Treatment of superior cervical ganglia neurons with pro-NGF resulted in cell death(Fig. 9D).
  • Treatment with mature NGF resulted in very little cell death (Fig. 9C).
  • Example 8 Expression of BDNF in eukaryotic cells:
  • Recombinant adenovirus may be utilized to express rat BDNF in 293T cells, or endothelial cells, an in vivo source of BDNF in the CNS and extraneuronal sites (Donovan, M. J., et al. 2000. Development 127:4531-4540; Leventhal, C, et al. 1999. Mol. Cell. Neurosci. 13:450-464). Forty-eight hours following cell infection, BDNF released into the culture media is detected by Western blot using an antibody specific for epitopes in the mature BDNF protein. Immunoreactive proteins of 30 kDa, 28 kDa, 24 kDa and 14 kDa are secreted into the media of 293 cells (Fig.
  • Example 9 Identification of proteases that are essential for the cleavage of a neurotrophin, BDNF, to its mature form:
  • plasmin, MMP-3 or MMP-7 can cleave secreted BDNF
  • recombinant proteases were added to the media from adenoviral infected cells, and all these enzymes led to a reduction in the proforms of 30 kDa in 293 cells while MMP-2 was without effect (Fig. 10A).
  • Fig. 10B In endothelial media, only plasmin and MMP-7 appeared to cleave the large proform (Fig. 10B), perhaps reflecting differences in TEVIP by endothelial cells as compared to 293 cells.
  • adenovirally infected cultures were incubated with plasmin, or a combination of plasmin plus aprotinin in the media for 48 hours prior to media collection.
  • inhibition of plasmin activity upon addition of aprotinin reduced the cleavage of high molecular weight forms of BDNF, strongly suggesting that these higher molecular weight proforms are released from cells, and that plasmin cleavage occurs on the surface of intact cells.
  • NGF native murine NGF
  • R-R (-1, 0) mutant to A-A constructs have been prepared. Because the NGF start site does not conform to a Kozak consensus sequence, impairing efficient translation, the 15 bases upstream of the NGF ATG (-14 to 0) was substituted for (-14 to 0) of the NT-3 sequence, yielding a cDNA which utilizes the NT-3 Kozak site and the entire NGF coding sequence.
  • Both native His-tagged NGF and the (R-R) to (A-A) mutant have been isolated from the media of transiently transfected 293 cells using a Ni-Sepharose column.
  • the His-tagged NGF migrating at 14 kDa, displays no loss of biological activity as compared to commercial recombinant NGF, as assessed in PC 12 neurite outgrowth assays.
  • Binding of cleavage resistant neurotrophins may be compared to native, mature NGF or BDNF to either p75, or their cognate trk receptors, using competition displacement studies. The methodology for these studies is well described in recent publications (Ryden, M., et al. 1997. The Journal of Biological Chemistry 272:16322-16328).
  • mature-His tagged or cleavage resistant-His tagged neurotrophin is radioiodinated to a specific activity of approximately 2800dpm/fmole. Iodinations using 0.5 ⁇ g of neurotrophin routinely generate material for approximately 20 binding studies (Hempstead, B. L., et al. 1991. Nature 350:678-683).
  • 3T3 cells stably expressing trk A or trk B, or 3T3 cells stably expressing p75 are incubated concomitantly with radioiodinated neurotrophin (at 10 "9 M) and unlabeled native neurotrophin at increasing concentrations (10 ⁇ 10 to 10 "8 M) at 4°C for 2 hours.
  • Bound ligand is separated from free by centrifugation of cells as described (Ryden M., et al., EMBO J. 14: 1979-90) and the binding properties of each mutant neurotrophin is determined by calculating the IC 50 (the concentration of inhibitor which reduces binding by 50%).
  • tissue expression of neurotrophins To determine the tissue expression of neurotrophins, adult mouse tissues were probed with antibodies that recognize the mature region of NGF or BNDF. In adult tissues, both mature (approximately 14 kD) and larger preproisoforms of both NGF and BDNF were detected (Fig. 11 A and B). The larger isoforms of NGF were also present in the commercial preparations of recombinant NGF. In the tissue extracts from adult mice, the detectable neurotrophin proteins ranged in size from 14 kD (mature form) to approximately 30 kD (size of the unprocessed, preproform). Intermediately sized proteins of 16, 18, and 22 kD for NGF and 18, 22, 28, and 30 kD for BDNF were also detected. The data demonstrates that proforms of NGF and BDNF are expressed at significant levels in many adult tissues. In addition, the data shows that tissues process NGF and BDNF differently.
  • Antibodies specific to the pro-domain of pro-BDNF and pro-NGF The antibodies available to date have been generated to the mature domain of the neurotrophins, thus recognizing both mature and pro-forms.
  • GST-fusion proteins encoding amino acids 20-80 of BDNF or amino acids 20-82 of NGF were generated and used as immunogens in chickens (for BDNF) or rabbits (for NGF).
  • the antisera was affinity purified using columns of the immunogen.
  • the antisera to pro-NGF detects pro-NGF, but not mature NGF (Fig. 12 A).
  • the antisera to pro-BDNF detcts pro-BDNF, but not mature BDNF (Fig. 12B).
  • Example 13 Pro-BDNF and pro-NGF are expressed in lesions of vascular injury:
  • Example 14 Expression of pro- and mature NGF and BDNF in human atherosclerotic lesions:
  • Example 15 Construction of a replication deficient recombinant adenovirus based on human serotype 5:
  • Neurotrophin cDNA is subcloned in the shuttle vector (p.shuttle.cmv) and cotransformed with pAdEasy (containing the adenovirus serotype 5 genome with deletions in E1-E3 regions) in bacteria. Homologous recombination replaces the El region of pAdEasy with the mutant neurotrophin cDNA and plasmids are isolated and screened for neurotrophin cDNA expression by PCR. Recombinant plasmid is transfected into 293 cells and viral particles are plaque purified. High titer stocks are prepared, using cesium chloride ultracentrifugation, and the viral concentration determined by plaque titration.
  • Viral stocks are used to infect 293 cells at a MOI of 10 for 90 min, and neurotrophins purified from the media 2 days after infection using Ni chromatography as outlined above. Purity is assessed by silver stain; if additional purification is necessary to obtain preparations which are >95% pure, reverse phase chromatography is performed (Ryden, M., et al. 1995. EMBO J 14:1979-90).
  • Trk activation is assessed in 3T3 cells expressing either trk A, trk B or trk C.
  • cells are treated with mature or cleavage resistant neurotrophins (10 "10 to 10 "8 M).
  • the rapid trk autophosphorylation (within 5 min), and pho ⁇ horylation and activation of well defined downstream signaling events such as MAP kinase and PI-3 kinase may be assessed by standard methods. From cells lysed in detergent buffer containing phosphatase and protease inhibitors, trk, or MAP kinase are immunoprecipitated and Western blotted using anti-PY antisera.
  • MAP and PI-3 kinase kinase activation is quantified using immunoprecipitation (IP)-kinase assays, as has been described (Hempstead, B. L., et al. 1992. Neuron 9:883-896).
  • IP immunoprecipitation
  • the dose response analysis of ligand binding to trk may then be compared with the induction of trk activation.
  • 293 cells transiently expressing p75 and TRAF6 may be used for dose- response analysis, by incubating cells with mature or cleavage resistant neurotrophin (10 "10 to 10 "8 M) for 5 minutes. Cells are then lysed, and a co-immunoprecipitation analysis of p75:TRAF6 complexes performed by immunoprecipitating with anti-p75 and probing for FLAG-tagged TRAF6. Based on prior studies (Kursigawa and Chao, 1999), the neurotrophins must be present at concentrations of 4 nM or greater to induce complex formation between p75 and TRAF6. Cleavage resistant neurotrophins which have enhanced binding affinity for p75 may be identified in this way.
  • Cleavage resistant mutants may also be characterized for binding and activation of p75 and trks independently and in cells which co-express both receptors.
  • the dose responsiveness of these cells to mature and cleavage resistant neurotrophins may be determined using trk autophosphorylation, MAP kinase activation and PI-3 kinase activation as trk signaling predominates when both receptors are expressed (Casaccia- Bonnefil, P., et al. 1999. The Functional Roles of Glial Cells in Health and Disease. Plenum, New York).
  • Example 17 Biological effects of cleavage resistant neurotrophins:
  • trk A or trk B expressing fibroblasts are used in cell proliferation analysis. These assays do not require p75 expression, and are highly quantitative (See Arevalo, J. C, et al. 2000. Mol Cell Biol 20:5908-16), thus allowing activation by native neurotrophins to be distinguished from those mutants which may exhibit reduced activity.
  • cells cultured in serum free media are treated with or without increasing concentrations of native or cleavage resistant neurotrophins for 24 hr, then pulsed with H-thymidine.
  • Thymidme incorporation as a measurement of cell proliferation, is quantitated in a filter binding assay (Arevalo, J. C, id.).
  • the cleavage resistant neurotrophins exhibit reduced proliferation when they are impaired in trk binding.
  • the first model is neurotrophin induced apoptosis of p75 expressing vascular smooth muscle cells (Wang, S., et al. 2000. Am J Pathol 157:1247-1258) which exhibit dose dependent increases in apoptosis using 2-5 nM NGF or BDNF, as quantitated by flow cytometric analysis of Annexin V binding. Concentrations of neurotrophin below 2 nM are not effective whereas dose responsiveness at 2-5nM concentrations provides a quantitative analysis necessary to distinguish between ligands with reduced or enhanced activation of p75.
  • p75 expressing vascular smooth muscle cells are treated with native or cleavage resistant neurotrophins for 18 hrs, and the proportion of cells exhibiting early or late apoptosis quantitated using flow cytometric analysis.
  • Cells with Annexin V binding, but no propidium iodide uptake (early apoptosis) or both Annexin V binding and propidium iodide uptake (indicative of late apoptosis) are quantitated readily, as has been detailed (Wang, S., et al. 2000. Am J Pathol 157:1247-1258).
  • rat neonatal oligodendrocyte assay may be used (Casaccia- Bonnefil, P., et al. 1996. Nature 383:716-719).
  • Cell cultured in oligodendrocyte differentiation media is switched to serum free media on neurotrophins (0.5 to 10 nM) and cell apoptosis is quantitated by TUNEL analysis at 8 hours.
  • neuronal cultures may be used to compare the biological actions of native or cleavage resistant neurotrophins on cells which express both trk and p75, and exhibit responsiveness by neuritogenesis and survival.
  • Primary cell cultures may be used, for example, cervical ganglion neurons, hippocampal neurons or dorsal root ganglion neurons.
  • stable PC 12 cell clones expressing either trk A or trk B at significant levels (approximately 70,000 receptors/cell) and have well characterized responses in terms of neurite extension (Hempstead, B. L., et al. 1992. Neuron 9:883- 896) are preferred.
  • Dose reponse studies (0.5 X 10 "11 M) are performed using native or cleavage resistant proneurotrophin and neurite outgrowth at 48 hours is quantitated by counting cells with neurites of greater than two cell bodies in length.
  • Cell survival assays may be performed, using neurons from rat dorsal root ganglia. These cells express either trk A and p75, or trk B and p75, and thus are useful in assessing the biological activities of mutant NGF or BDNF. Results obtained using cells which co-express both p75 and trk receptors are interpreted in the context of those obtained using cells which express each receptor independently, as well as in the results of receptor binding studies.
EP02729305A 2001-05-25 2002-05-24 LIGAND A FORTE AFFINITE POUR LE RECEPTEUR DE LA NEUROTROPHINE p75 Expired - Lifetime EP1575477B1 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US29382301P 2001-05-25 2001-05-25
US293823P 2001-05-25
US30551001P 2001-07-13 2001-07-13
US305510P 2001-07-13
PCT/US2002/016540 WO2002096356A2 (fr) 2001-05-25 2002-05-24 Ligand a forte affinite pour le recepteur de la neurotrophine p75

Publications (3)

Publication Number Publication Date
EP1575477A2 true EP1575477A2 (fr) 2005-09-21
EP1575477A4 EP1575477A4 (fr) 2006-11-08
EP1575477B1 EP1575477B1 (fr) 2012-04-25

Family

ID=26968166

Family Applications (1)

Application Number Title Priority Date Filing Date
EP02729305A Expired - Lifetime EP1575477B1 (fr) 2001-05-25 2002-05-24 LIGAND A FORTE AFFINITE POUR LE RECEPTEUR DE LA NEUROTROPHINE p75

Country Status (7)

Country Link
US (2) US7507799B2 (fr)
EP (1) EP1575477B1 (fr)
AT (1) ATE554784T1 (fr)
AU (1) AU2002259305A1 (fr)
CA (1) CA2447986A1 (fr)
DK (1) DK1575477T3 (fr)
WO (1) WO2002096356A2 (fr)

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2002259305A1 (en) * 2001-05-25 2002-12-09 Cornell Research Foundation, Inc. High affinity ligand for p75 neurotrophin receptor
US8066997B2 (en) 2002-12-20 2011-11-29 Anders Nykjaer Modulation of activity of neurotrophins
DE10317369A1 (de) * 2003-04-15 2004-11-11 Scil Proteins Gmbh proNGF als pharmazeutisch wirksames Mittel zur Behandlung demyelinisierender Erkrankungen
EP1500399A1 (fr) * 2003-07-24 2005-01-26 Institut Pasteur Immunisation active et passive contre neurotrophines pro-apoptotiques pour le traitement ou la prévention des maladies neurologiques
CN1897977A (zh) * 2003-10-20 2007-01-17 Ns基因公司 帕金森氏病的体内基因治疗
EP1682170A2 (fr) * 2003-11-07 2006-07-26 Lay Line Genomics SpA Compositions pouvant empecher des troubles neurodegeneratifs et procede pour tester l'activite de telles compositions
ITRM20030601A1 (it) 2003-12-24 2005-06-25 Lay Line Genomics Spa Metodo per l'umanizzazione di anticorpi e anticorpi umanizzati con esso ottenuti.
ITRM20050045A1 (it) * 2005-02-02 2006-08-03 Consiglio Nazionale Ricerche Proteina isolata da eisenia foetida simile all'ngf umano e usi relativi.
ITRM20050290A1 (it) * 2005-06-07 2006-12-08 Lay Line Genomics Spa Uso di molecole in grado di inibire il legame tra ngf e il suo recettore trka come analgesici ad effetto prolungato.
US20080050776A1 (en) * 2006-05-26 2008-02-28 Kenneth Neet Stable mutated pro nerve growth factors
HUE048781T2 (hu) 2006-12-21 2020-08-28 H Lundbeck As Proneurotrofinok aktivitásának modulációja
JP5871249B2 (ja) * 2010-05-31 2016-03-01 国立研究開発法人産業技術総合研究所 成長因子のプロペプチド
US20130115222A1 (en) * 2010-06-15 2013-05-09 Cornell University Methods of limiting microvascular damage following acute myocardial ischemia
US20130336988A1 (en) * 2010-11-17 2013-12-19 New York University Methods for treating early stage or mild neurological disorders
US11214610B2 (en) 2010-12-01 2022-01-04 H. Lundbeck A/S High-purity production of multi-subunit proteins such as antibodies in transformed microbes such as Pichia pastoris
US9067988B2 (en) 2010-12-01 2015-06-30 Alderbio Holdings Llc Methods of preventing or treating pain using anti-NGF antibodies
AU2011336470B8 (en) 2010-12-01 2017-09-14 Alderbio Holdings Llc Anti-NGF compositions and use thereof
US9078878B2 (en) 2010-12-01 2015-07-14 Alderbio Holdings Llc Anti-NGF antibodies that selectively inhibit the association of NGF with TrkA, without affecting the association of NGF with p75
US9884909B2 (en) 2010-12-01 2018-02-06 Alderbio Holdings Llc Anti-NGF compositions and use thereof
US9539324B2 (en) 2010-12-01 2017-01-10 Alderbio Holdings, Llc Methods of preventing inflammation and treating pain using anti-NGF compositions
RU2486918C1 (ru) * 2011-10-25 2013-07-10 Федеральное государственное бюджетное образовательное учреждение высшего профессионального образования "Московский государственный университет имени М.В. Ломоносова" (МГУ) Способ стимулирования восстановления периферической иннервации тканей с помощью векторных конструкций
MX353074B (es) 2011-12-19 2017-12-19 Wacker Chemie Ag Mutantes profactor de crecimiento del nervio (ngf) novedosos y usos de los mismos en la produccion de factor de crecimiento del nervio beta.
US20140256639A1 (en) * 2013-02-14 2014-09-11 The Regents Of The University Of California Peptoid neutralizing agents
US20160220639A1 (en) * 2013-09-11 2016-08-04 New York University Methods and compositions for treating bone diseases
CN104774264B (zh) * 2014-01-15 2018-09-14 上海易乐生物技术有限公司 抗人proBDNF单克隆抗体及其在疼痛中的作用
US10500273B2 (en) 2015-03-02 2019-12-10 180 Therapeutics Lp Method of treating a localized fibrotic disorder using an IL-33 antagonist
US20210079050A1 (en) * 2018-02-20 2021-03-18 Carmel Haifa University Economic Corporation Ltd. Compositions and methods for delivery and expression of small inhibitory peptides and use thereof
IT202000007720A1 (it) * 2020-04-10 2021-10-10 Alessandra Marconi Peptidi e loro usi
IT202000032423A1 (it) * 2020-12-24 2022-06-24 Univ Degli Studi Di Trieste Metodo per la produzione di forme processate proteoliticamente di fattori trofici o fattori di crescita
WO2022226170A1 (fr) * 2021-04-22 2022-10-27 Cornell University Ligand immunomodulateur b7-1 médiant le remodelage synaptique au moyen de p75ntr

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997036607A1 (fr) * 1996-03-29 1997-10-09 The Regents Of The University Of California Derives de peptides de synthese presentant une activite neurotrophique de type facteur de croissance nerveuse
WO2000075278A2 (fr) * 1999-06-07 2000-12-14 The Trustees Of Columbia University In The City Of New York Gene codant pour 'nade' ou activateur de mort cellulaire associe a p75ntr et procede de son utilisation
WO2001052843A1 (fr) * 2000-01-18 2001-07-26 Mcgill University Composes cycliques peptidomimetiques a coude beta

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5169762A (en) * 1983-03-03 1992-12-08 Genentech, Inc. Human nerve growth factor by recombinant technology
US5169764A (en) * 1990-08-08 1992-12-08 Regeneron Pharmaceuticals, Inc. Multitrophic and multifunctional chimeric neurotrophic factors, and nucleic acids and plasmids encoding the chimeras
JPH10212241A (ja) * 1996-05-27 1998-08-11 Sumitomo Pharmaceut Co Ltd Bdnfを安定に含有する製剤
US5840736A (en) * 1996-11-13 1998-11-24 Vertex Pharmaceuticals Incorporated Methods and compositions for stimulating neurite growth
WO2000024415A2 (fr) 1998-10-28 2000-05-04 Cornell Research Foundation, Inc. Methodes de regulation de l'angiogenese et de l'integrite vasculaire a l'aide de ligands des recepteurs trk
GB0007918D0 (en) 2000-03-31 2000-05-17 Npower Passive valve assembly
AU2002259305A1 (en) 2001-05-25 2002-12-09 Cornell Research Foundation, Inc. High affinity ligand for p75 neurotrophin receptor

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997036607A1 (fr) * 1996-03-29 1997-10-09 The Regents Of The University Of California Derives de peptides de synthese presentant une activite neurotrophique de type facteur de croissance nerveuse
WO2000075278A2 (fr) * 1999-06-07 2000-12-14 The Trustees Of Columbia University In The City Of New York Gene codant pour 'nade' ou activateur de mort cellulaire associe a p75ntr et procede de son utilisation
WO2001052843A1 (fr) * 2000-01-18 2001-07-26 Mcgill University Composes cycliques peptidomimetiques a coude beta

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
CORTAZZO MH ET AL.: "Nerve Growth Factor (NGF)-Mediated Protection of Neural Crest Cells from Antimitotic Agent-Induced Apoptosis: The Role of the Low-Affinity NGF Receptor" THE JOURNAL OF NEUROSCIENCE, vol. 16, no. 12, 15 June 1996 (1996-06-15), pages 3895-3899, XP002400288 ISSN: 0270-6474 *
GENTRY JJ ET AL.: "Nerve Growth Factor Activation of Nuclear Factor .kappa.B through Its p75 Receptor Is an Anti-apoptotic Signal in RN22 Schwannoma Cells" THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 275, no. 11, 17 March 2000 (2000-03-17), pages 7558-7565, XP002400289 ISSN: 0021-9258 *
LEE R ET AL: "Regulation of Cell Survival by Secreted Proneurotrophins", 30 November 2001 (2001-11-30), SCIENCE, AMERICAN ASSOCIATION FOR THE ADVANCEMENT OF SCIENCE, US, WASHINGTON, DC, PAGE(S) 1945 - 1948, XP003002960, ISSN: 0036-8075 *
NYKJAER A ET AL: "Sortilin is essential for proNGF-induced neuronal cell death" NATURE, NATURE PUBLISHING GROUP, LONDON, GB, vol. 427, 26 February 2004 (2004-02-26), pages 843-848, XP002286438 ISSN: 0028-0836 *
See also references of WO02096356A2 *
SOILU-HAENNINEN M ET AL: "Nerve growth factor signaling through p75 induces apoptosis in Schwann cells via a Bcl-2-independent pathway" JOURNAL OF NEUROSCIENCE, NEW YORK, NY, US, vol. 19, no. 12, 15 June 1999 (1999-06-15), pages 4828-4838, XP002234198 ISSN: 0270-6474 *

Also Published As

Publication number Publication date
US20080025978A1 (en) 2008-01-31
DK1575477T3 (da) 2012-07-23
WO2002096356A3 (fr) 2006-05-18
CA2447986A1 (fr) 2002-12-05
US8034347B2 (en) 2011-10-11
EP1575477A4 (fr) 2006-11-08
ATE554784T1 (de) 2012-05-15
AU2002259305A1 (en) 2002-12-09
AU2002259305A8 (en) 2006-11-02
EP1575477B1 (fr) 2012-04-25
WO2002096356A2 (fr) 2002-12-05
US20030087804A1 (en) 2003-05-08
US7507799B2 (en) 2009-03-24

Similar Documents

Publication Publication Date Title
US8034347B2 (en) Method for inhibiting apoptosis through the p75 neurotrophin receptor
RU2146262C1 (ru) Пептиды, способ их получения, фармацевтическая композиция и способ ее получения
JP4685452B2 (ja) Actriib融合ポリペプチドおよびその使用
EP1124846B1 (fr) Sequences nucleotidiques et proteiques de gene nogo et procedes reposant sur ces sequences
US20120196803A1 (en) Fusion proteins for delivery of gdnf and bdnf to the central nervous system
MXPA05002968A (es) Activacion de miostatina por metaloproteasa, y metodos de modular la actividad de miostatina.
CA2859412C (fr) Distribution d'agents therapeutiques employant des segments polypeptidesliant un collagene bacterien
AU1703595A (en) Therapeutic use of myelin-associated glycoprotein (mag)
JPH11508886A (ja) ミエリン会合糖タンパク質(mag)およびそのインヒビターを用いた組成物および方法
WO1998022499A2 (fr) Systeme de regulation de la croissance tumorale neuronale et neurale, anticorps destines a cet effet et utilisations de ceux-ci
KR20010041418A (ko) 프로테아제-활성화되는 수용체 4 및 그것의 사용
US7772204B1 (en) Perlecan and growth factor for wound and cutaneous injury healing
WO2003000281A1 (fr) Neuroprotection et/ou neurorestoration via le recepteur de l'activine neurale de type iib
US20040203102A1 (en) Antiplasmin cleaving enzyme
US5578566A (en) KGF receptor-derived antagonists of KGF binding
CA2144515A1 (fr) Traitement des ulceres gastro-intestinaux
CA2107475A1 (fr) Proteine fixatrice du facteur de croissance apparente a l'insuline isole du tissu osseux humain
KR20180099092A (ko) β1 인테그린 신호전달 억제 활성을 갖는 펩타이드 및 이를 포함하는 약학 조성물
US6329500B1 (en) Transforming growth factor-β binding site
CA2368937A1 (fr) Prevention de lesions cerebrales liees a des accidents vasculaires cerebraux
WO2001089450A2 (fr) Traitement de pathologies musculosquelettiques par le polypeptide lp85 et des analogues de celui-ci
WO1995015176A1 (fr) Procedes activant la duree de vie et facilitant la differentiation des sous-classes de neurones cholinergiques et serotonergiques a l'aide du facteur 5 de croissance des fibroblastes
JPH1099084A (ja) 新規タンパク質およびそのdna
JP2003079376A (ja) 新規なジンクフィンガータンパク質ezi及びその遺伝子
JP2003079373A (ja) 新規なジンクフィンガータンパク質ezi及びその遺伝子

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20031229

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

PUAK Availability of information related to the publication of the international search report

Free format text: ORIGINAL CODE: 0009015

RIC1 Information provided on ipc code assigned before grant

Ipc: C12N 15/00 20060101ALI20060707BHEP

Ipc: A01N 61/00 20060101ALI20060707BHEP

Ipc: C07K 14/00 20060101AFI20060707BHEP

Ipc: A01N 37/18 20060101ALI20060707BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20061006

17Q First examination report despatched

Effective date: 20080514

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 60242745

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: A61K0006000000

Ipc: A61K0038070000

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 38/10 20060101ALI20110616BHEP

Ipc: A61K 38/07 20060101AFI20110616BHEP

Ipc: A61K 38/48 20060101ALI20110616BHEP

Ipc: A61K 38/08 20060101ALI20110616BHEP

Ipc: A61K 38/55 20060101ALI20110616BHEP

Ipc: A61K 38/12 20060101ALI20110616BHEP

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

DAX Request for extension of the european patent (deleted)
GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 554784

Country of ref document: AT

Kind code of ref document: T

Effective date: 20120515

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 60242745

Country of ref document: DE

Effective date: 20120621

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

REG Reference to a national code

Ref country code: NL

Ref legal event code: VDEP

Effective date: 20120425

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 554784

Country of ref document: AT

Kind code of ref document: T

Effective date: 20120425

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120425

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120425

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120425

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120726

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120827

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20120531

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120425

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20120531

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20120531

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120425

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120425

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120425

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

Effective date: 20130131

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 60242745

Country of ref document: DE

Effective date: 20121201

26N No opposition filed

Effective date: 20130128

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20120625

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120805

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20120524

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20121201

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120425

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20120524

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20140527

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DK

Payment date: 20140602

Year of fee payment: 13

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20150531

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20150524

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150531

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150524